TWI570240B - A conditional replicating cytomegalovirus as a vaccine for cmv - Google Patents

A conditional replicating cytomegalovirus as a vaccine for cmv Download PDF

Info

Publication number
TWI570240B
TWI570240B TW101132064A TW101132064A TWI570240B TW I570240 B TWI570240 B TW I570240B TW 101132064 A TW101132064 A TW 101132064A TW 101132064 A TW101132064 A TW 101132064A TW I570240 B TWI570240 B TW I570240B
Authority
TW
Taiwan
Prior art keywords
cmv
virus
shield
protein
cells
Prior art date
Application number
TW101132064A
Other languages
Chinese (zh)
Other versions
TW201311898A (en
Inventor
傅董民
王岱
慕尼斯沃拉 巴布 麥迪
Original Assignee
默沙東公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=47832753&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=TWI570240(B) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by 默沙東公司 filed Critical 默沙東公司
Publication of TW201311898A publication Critical patent/TW201311898A/en
Application granted granted Critical
Publication of TWI570240B publication Critical patent/TWI570240B/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • C07K14/01DNA viruses
    • C07K14/03Herpetoviridae, e.g. pseudorabies virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/245Herpetoviridae, e.g. herpes simplex virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • C07K14/01DNA viruses
    • C07K14/03Herpetoviridae, e.g. pseudorabies virus
    • C07K14/04Varicella-zoster virus
    • C07K14/045Cytomegalovirus
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5252Virus inactivated (killed)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5254Virus avirulent or attenuated
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55505Inorganic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55577Saponins; Quil A; QS21; ISCOMS
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/04Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with deoxyribosyl as saccharide radical
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16111Cytomegalovirus, e.g. human herpesvirus 5
    • C12N2710/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biophysics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Description

作為細胞巨大病毒疫苗之條件式複製CMV Conditional replication CMV as a cellular giant virus vaccine

本發明係關於使用經遺傳修飾之條件式複製缺陷型細胞巨大病毒(CMV)誘導對CMV之免疫反應的方法。本發明亦係關於CMV,其已經重組改變以允許外部控制病毒複製。本發明亦涵蓋包含該複製缺陷型CMV之組合物。 The present invention relates to a method for inducing an immune response to CMV using genetically modified conditionally replicating defective cell giant virus (CMV). The invention also relates to CMVs that have been recombined to allow external control of viral replication. Compositions comprising the replication defective CMV are also contemplated by the present invention.

細胞巨大病毒(CMV)(亦稱為人類皰疹病毒5,HHV-5)係歸類為皰疹病毒科(herpesviridae)β亞科之成員之皰疹病毒。根據疾病控制與預防中心(Centers for Disease Control and Prevention),在人類群體中相當廣泛地發現CMV感染,估計美國成人群體中有40-80%已受到感染。該病毒主要經由體液傳播且經常自懷孕母親傳遞至胎兒或新生兒。在大多數個體中,CMV感染係潛伏的,但病毒活化可導致高燒、發冷、疲乏、頭痛、噁心及脾腫大。 The cell giant virus (CMV) (also known as human herpesvirus 5, HHV-5) is a herpes virus classified as a member of the beta subfamily of herpesviridae. According to the Centers for Disease Control and Prevention, CMV infection is fairly widespread in the human population, with an estimated 40-80% of the adult population in the United States being infected. The virus is primarily transmitted through body fluids and is often delivered from a pregnant mother to a fetus or newborn. In most individuals, CMV infection is latent, but viral activation can lead to high fever, chills, fatigue, headache, nausea, and splenomegaly.

儘管多數人類CMV感染沒有症狀,但免疫受損個體(例如HIV陽性患者、同種異體移植患者及癌症患者)或免疫系統尚未發育完全者(例如新生兒)之CMV感染可尤其成問題(Mocarski等人,Cytomegalovirus,in Field Virology,2701-2772,編輯:Knipes及Howley,2007)。此等個體之CMV感染可造成嚴重病狀,包括肺炎、肝炎、腦炎、結腸炎、葡萄膜炎、視網膜炎、失明及神經病變,以及其他有害病況。另外,懷孕期間之CMV感染係出生缺陷之主要原因(Adler,2008 J.Clin Virol,41:231;Arvin等人,2004 Clin Infect Dis,39:233;Revello等人,2008 J Med Virol,80:1415)。CMV感染各種活體內細胞,包括單核球、巨噬細胞、樹突細胞、嗜中性球、內皮細胞、上皮細胞、纖維母細胞、神經元、平滑肌細胞、肝細胞及基質細胞(Plachter等人1996,Adv.Virus Res.46:195)。儘管臨床CMV分離物在多種細胞類型中複製,但實驗室株AD 169(Elek及Stern,1974,Lancet 1:1)及Towne(Plotkin等人,1975,Infect.Immun.12:521)幾乎僅在纖維母細胞中複製(Hahn等人,2004,J.Virol.78:10023)。規定源於病毒在纖維母細胞中之連續傳代及最終適應之向性限制為衰減標記物(Gerna等人,2005,J.Gen.Virol.86:275;Gerna等人,2002,J.Gen Virol.83:1993;Gerna等人,2003,J.Gen Virol.84:1431;Dargan等人,2010,J.Gen Virol.91:1535)。已將造成人類CMV實驗室株中上皮細胞、內皮細胞、白血球及樹突細胞向性損失之突變作圖至三個開放閱讀框(ORF):UL128、UL130及UL131(Hahn等人,2004,J.Virol.78:10023;Wang及Shenk,2005J.Virol.79:10330;Wang及Shenk,2005 Proc Natl Acad Sci USA.102:18153)。生化及重構研究顯示,UL128、UL130及UL131組裝至gH/gL支架上以形成五聚gH複合物(Wang及Shenk,2005 Proc Natl Acad Sci USA.102:1815;Ryckman等人,2008 J.Virol.82:60)。在病毒粒子中恢復此複合物可恢復實驗室株中之病毒上皮向性(Wang及Shenk,2005 J.Virol.79:10330)。 Although most human CMV infections are asymptomatic, CMV infection in immunocompromised individuals (such as HIV-positive patients, allograft patients, and cancer patients) or those whose immune system is not fully developed (eg, neonates) can be particularly problematic (Mocarski et al. , Cytomegalovirus, in Field Virology, 2701-2772, edited by Knipes and Howley, 2007). CMV infection in these individuals can cause serious conditions including pneumonia, hepatitis, encephalitis, colitis, uveitis, retinitis, blindness and neuropathy, as well as other harmful conditions. In addition, CMV infection during pregnancy is the main cause of birth defects (Adler, 2008 J. Clin Virol, 41: 231; Arvin et al., 2004 Clin Infect Dis, 39: 233; Revello et al., 2008 J Med Virol, 80: 1415). CMV infects a variety of living cells, including mononuclear cells, macrophages, dendritic cells, neutrophils, endothelial cells, epithelial cells, fibroblasts, neurons, smooth muscle cells, hepatocytes, and stromal cells (Plachter et al. 1996, Adv. Virus Res. 46: 195). Although clinical CMV isolates replicated in a variety of cell types, laboratory strains AD 169 (Elek and Stern, 1974, Lancet 1:1) and Towne (Plotkin et al., 1975, Infect. Immun. 12: 521) are almost exclusively Replication in fibroblasts (Hahn et al., 2004, J. Virol. 78: 10023). It is stipulated that the continuation of continuous passage and final adaptation of the virus in fibroblasts is limited to attenuation markers (Gerna et al., 2005, J. Gen. Virol. 86: 275; Gerna et al., 2002, J. Gen. Virol. 83: 1993; Gerna et al, 2003, J. Gen Virol. 84: 1431; Dargan et al, 2010, J. Gen Virol. 91:1535). Mutations in the tropism of epithelial cells, endothelial cells, white blood cells and dendritic cells in human CMV laboratory strains have been mapped to three open reading frames (ORF): UL128, UL130 and UL131 (Hahn et al., 2004, J). .Virol. 78:10023; Wang and Shenk, 2005 J. Virol. 79: 10330; Wang and Shenk, 2005 Proc Natl Acad Sci USA. 102: 18153). Biochemical and remodeling studies have shown that UL128, UL130 and UL131 are assembled onto gH/gL scaffolds to form pentameric gH complexes (Wang and Shenk, 2005 Proc Natl Acad Sci USA. 102: 1815; Ryckman et al., 2008 J. Virol) .82:60). Recovery of this complex in virions restored viral epithelial tropism in laboratory strains (Wang and Shenk, 2005 J. Virol. 79: 10330).

已懷疑內皮及上皮向性損失係先前評估為諸如Towne等疫苗之缺陷(Gerna等人,2002,J.Gen Virol.83:1993;Gerna等人,2003,J.Gen Virol.84:1431)。感染天然CMV之人類個體之血清中之中和抗體針對病毒上皮進入之活性係針對纖維母細胞進入之15倍(Cui等人,2008 Vaccine 26:5760)。具有原發性感染之人類快速地產生針對病毒內皮及上皮進入之中和抗體,但僅緩慢地產生針對病毒纖維母細胞進入之中和抗體(Gerna等人,2008 J.Gen.Virol.89:853)。此外,在接受Towne疫苗之人類個體之免疫血清中不存在針對病毒上皮及內皮進入之中和活性(Cui等人,2008 Vaccine 26:5760)。最近,闡述一組來自四名具有HCMV感染之供體之人類單株抗體,且來自該組之中和效能較高之純系識別五聚gH複合物抗原(Macagno等人,2010 J.Virol.84:1005)。 Endothelial and epithelial tropism has been previously assessed as a defect in vaccines such as Towne (Gerna et al, 2002, J. Gen Virol. 83: 1993; Gerna et al, 2003, J. Gen Virol. 84: 1431). In the serum of human subjects infected with natural CMV, the activity of neutralizing antibodies against viral epithelial entry was 15 times that of fibroblasts (Cui et al, 2008 Vaccine 26: 5760). Humans with primary infections rapidly develop neutralizing antibodies against the viral endothelium and epithelium, but only slowly produce neutralizing antibodies against viral fibroblasts (Gerna et al., 2008 J. Gen. Virol. 89: 853). In addition, there is no neutralizing activity against viral epithelium and endothelium in the immune sera of human subjects receiving the Towne vaccine (Cui et al., 2008 Vaccine 26: 5760). Recently, a panel of human monoclonal antibodies from four donors with HCMV infection was described, and from this group, the homologous recognition of the pentameric gH complex antigen was high (Macagno et al., 2010 J. Virol. 84 :1005).

本發明係關於條件式複製缺陷型CMV(rdCMV)及rdCMV在治療患者之CMV感染或與此一感染相關之病狀及/或降低其可能性之組合物及方法中的用途。本文所述rdCMV包含編碼一或多種融合蛋白之核酸,該等蛋白包含融合至去穩定蛋白之必需蛋白。在不存在穩定劑下,融合蛋白降解。因此,rdCMV可在允許複製之條件下(即,在存在穩定劑下)在組織培養物中生長,但在投與患者時(在不存在穩定劑下),減少且較佳防止複製。 The present invention relates to the use of conditional replication-deficient CMV (rdCMV) and rdCMV in compositions and methods for treating a CMV infection in a patient or a condition associated with such an infection and/or reducing the likelihood thereof. The rdCMVs described herein comprise nucleic acids encoding one or more fusion proteins comprising essential proteins fused to a destabilizing protein. The fusion protein degrades in the absence of a stabilizer. Thus, rdCMV can be grown in tissue culture under conditions that permit replication (i.e., in the presence of a stabilizer), but reduces and preferably prevents replication when administered to a patient (in the absence of a stabilizer).

本發明之一個實施例係條件式複製缺陷型CMV。rdCMV 包含編碼一或多種融合蛋白之核酸,該等蛋白包含融合至去穩定蛋白之必需蛋白。rdCMV中不再存在編碼野生型必需蛋白之核酸且因此病毒複製需要該融合蛋白。在一較佳實施例中,必需蛋白係選自IE1/2、UL51、UL52、UL79及UL84組成之群且去穩定蛋白係FKBP或其衍生物。 One embodiment of the invention is a conditional replication defective CMV. rdCMV A nucleic acid encoding one or more fusion proteins comprising an essential protein fused to a destabilizing protein is included. A nucleic acid encoding a wild-type essential protein is no longer present in rdCMV and thus the fusion protein is required for viral replication. In a preferred embodiment, the essential protein is selected from the group consisting of IE1/2, UL51, UL52, UL79, and UL84 and the destabilizing protein line FKBP or a derivative thereof.

本發明之另一實施例係包含經分離rdCMV及醫藥上可接受之載劑之組合物。該組合物可進一步包含佐劑,包括(但不限於)ISCOMATRIX®佐劑及磷酸鋁佐劑。 Another embodiment of the invention is a composition comprising isolated rdCMV and a pharmaceutically acceptable carrier. The composition may further comprise an adjuvant including, but not limited to, an ISCOMATRIX® adjuvant and an aluminum phosphate adjuvant.

本發明之另一實施例係rdCMV組合物在患者中誘導針對CMV之免疫反應的用途。可藉由投與本發明rdCMV來預防性或治療性治療患者。預防性治療提供足夠保護性免疫力以降低CMV感染之可能性或嚴重程度,該感染包括原發性感染、復發性感染(即,彼等源於潛伏CMV再活化者)及再感染(即,彼等源於與患者先前所經歷者不同株CMV之感染者)。在具體實施例中,給育齡女性、尤其早期青春期女性接種疫苗以降低在懷孕期間CMV感染(原發性、復發性或再)之可能性且因此降低將CMV傳染至胎兒之可能性。可執行治療性治療以降低現行CMV感染之時長/嚴重程度。 Another embodiment of the invention is the use of a rdCMV composition to induce an immune response against CMV in a patient. The patient can be treated prophylactically or therapeutically by administering the rdCMV of the invention. Prophylactic treatment provides sufficient protective immunity to reduce the likelihood or severity of CMV infection, including primary infections, recurrent infections (ie, those from latent CMV reactivation), and reinfection (ie, They originate from those infected with CMV different from those previously experienced by the patient). In a specific embodiment, women of childbearing age, particularly early adolescent women, are vaccinated to reduce the likelihood of CMV infection (primary, relapsing or re-) during pregnancy and thus reduce the likelihood of infecting CMV to the fetus. Therapeutic treatment can be performed to reduce the duration/severity of current CMV infections.

本發明之另一實施例係製備本發明rdCMV之方法,其包含在Shield-1存在下在上皮細胞(例如ARPE-19細胞,ATCC登錄號CRL-2302)上繁殖rdCMV。在一些實施例中,在微載體或其他高密度細胞培養系統上在上皮細胞繁殖rdCMV。 Another embodiment of the invention is a method of making a rdCMV of the invention comprising propagating rdCMV on epithelial cells (e.g., ARPE-19 cells, ATCC Accession No. CRL-2302) in the presence of Shield-1. In some embodiments, rdCMV is propagated in epithelial cells on a microcarrier or other high density cell culture system.

本發明係關於條件式複製缺陷型CMV(rdCMV)及rdCMV在治療患者之CMV感染或與此一感染相關之病狀及/或降低其可能性之組合物及方法中的用途。本文所述rdCMV編碼一或多種融合蛋白,該等融合蛋白包含融合至去穩定蛋白之必需蛋白,而非野生型必需蛋白。在不存在穩定劑下,融合蛋白由宿主細胞機器降解。在存在穩定劑下,融合蛋白穩定且不會降解。 The present invention relates to the use of conditional replication-deficient CMV (rdCMV) and rdCMV in compositions and methods for treating a CMV infection in a patient or a condition associated with such an infection and/or reducing the likelihood thereof. The rdCMV described herein encodes one or more fusion proteins comprising an essential protein fused to a destabilizing protein, rather than a wild type essential protein. In the absence of a stabilizer, the fusion protein is degraded by the host cell machinery. In the presence of a stabilizer, the fusion protein is stable and does not degrade.

用於本發明中之適宜融合蛋白在存在穩定劑下保留足夠必需蛋白活性以促進在宿主細胞中之病毒複製,且在不存在穩定劑下使CMV複製減少(較佳大於50%、75%、90%、95%或99%減少)。較佳地,用於融合蛋白中之必需蛋白編碼非結構蛋白且因此不會封裝至rdCMV病毒粒子中。本文所鑑別之適宜必需蛋白包括藉由必需基因IE1/2、UL51、UL52、UL79及UL84編碼之CMV蛋白。 Suitable fusion proteins for use in the present invention retain sufficient essential protein activity in the presence of a stabilizer to promote viral replication in a host cell and reduce CMV replication in the absence of a stabilizer (preferably greater than 50%, 75%, 90%, 95% or 99% reduction). Preferably, the essential protein used in the fusion protein encodes a non-structural protein and is therefore not encapsulated into the rdCMV virion. Suitable essential proteins identified herein include CMV proteins encoded by the essential genes IE1/2, UL51, UL52, UL79, and UL84.

去穩定蛋白及穩定劑之實例闡述於美國專利公開案2009/0215169中,該案件揭示使用小分子調節蛋白之穩定性的組合物、系統及方法。簡言之,將蛋白融合至影響穩定性之蛋白FKBP或其衍生物。外源添加之可透過細胞之小分子Shield-1(Shld-1)與FKBP或其衍生物相互作用並穩定融合蛋白。在Shield-1不存在下,FKBP或其衍生物引導融合蛋白由宿主細胞機器降解。 Examples of destabilizing proteins and stabilizers are set forth in U.S. Patent Publication No. 2009/0215169, which discloses compositions, systems and methods for the use of small molecule regulatory proteins. Briefly, the protein is fused to a protein FKBP or a derivative thereof that affects stability. The exogenously added cell-permeable small molecule Shield-1 (Shld-1) interacts with FKBP or its derivatives and stabilizes the fusion protein. In the absence of Shield-1, FKBP or its derivatives direct the fusion protein to be degraded by the host cell machinery.

在本發明實施例中,將必需CMV蛋白融合至FKBP或其衍生物。在Shield-1存在下,融合蛋白係穩定的。然而, 在Shield-1不存在下,FKBP或其衍生物引導融合蛋白由宿主細胞機器降解。 In an embodiment of the invention, the essential CMV protein is fused to FKBP or a derivative thereof. The fusion protein is stable in the presence of Shield-1. however, In the absence of Shield-1, FKBP or its derivatives direct the fusion protein to be degraded by the host cell machinery.

在不存在融合蛋白下,減少(與不含有去穩定必需蛋白之CMV相比,較佳大於50%、75%、90%、95%或99%)或防止rdCMV複製。 In the absence of the fusion protein, it is reduced (more preferably greater than 50%, 75%, 90%, 95% or 99% compared to CMV without the destabilizing essential protein) or prevents rdCMV replication.

欲用於本發明方法中之重組病毒亦在其病毒粒子上展示免疫原性五聚gH複合物。 The recombinant virus to be used in the method of the invention also displays an immunogenic pentameric gH complex on its virions.

實施例亦包括本文所述重組CMV或其組合物或包含該等CMV或組合物或由其組成之疫苗,其用於以下用途:(i)用於(a)療法(例如,人體);(b)藥物;(c)抑制CMV複製;(d)治療或預防CMV感染;或(e)治療、預防CMV相關疾病或延遲其發作或進展,(ii)用作用於以上各項之藥劑,或(iii)用於製備用於以上各項之藥劑。在該等用途中,重組CMV、其組合物及/或包含該等CMV或組合物或由其組成之疫苗可視情況與一或多種抗病毒劑(例如,抗病毒化合物或抗病毒免疫球蛋白;組合疫苗,如下文所述)組合使用。 Embodiments also include recombinant CMVs or compositions thereof, or vaccines comprising or consisting of the CMVs described herein, for use in (i) for (a) therapy (eg, human); b) drugs; (c) inhibiting CMV replication; (d) treating or preventing CMV infection; or (e) treating or preventing CMV-related diseases or delaying their onset or progression, (ii) acting as an agent for the above, or (iii) for the preparation of a medicament for use in the above. In such uses, the recombinant CMV, its compositions, and/or vaccines comprising or consisting of such CMVs or compositions may optionally be combined with one or more antiviral agents (eg, antiviral compounds or antiviral immunoglobulins; Combination vaccines, as described below) are used in combination.

本文所用「誘導免疫反應」係指條件式複製缺陷型CMV在所投與患者、較佳哺乳動物、更佳人類中產生免疫反應之能力,其中該反應包括(但不限於)產生特異性結合且較佳中和CMV及/或使T細胞活化之成份(例如抗體)。「保護性免疫反應」係降低患者感染CMV(包括原發性、復發性及/或再感染)之可能性及/或改善至少一種與CMV感染相關之病狀及/或降低CMV感染之嚴重程度/時長的免疫反應。 As used herein, "inducing an immune response" refers to the ability of a conditional replication-deficient CMV to produce an immune response in a patient, preferably a mammal, or a better human, wherein the response includes, but is not limited to, the production of specific binding. Preferably, the CMV and/or components (eg, antibodies) that activate T cells are neutralized. "Protective immune response" reduces the likelihood of a patient being infected with CMV (including primary, recurrent, and/or reinfection) and/or improves at least one condition associated with CMV infection and/or reduces the severity of CMV infection. / Duration of the immune response.

本文所用「免疫有效量」係指免疫原在投與患者時可誘導針對CMV之免疫反應之量,該免疫反應可保護患者免受CMV感染(包括原發性、復發性及/或再感染)及/或改善至少一種與CMV感染相關之病狀及/或降低患者之CMV感染之嚴重程度/時長。該量應足以顯著降低CMV感染之可能性或嚴重程度。可使用業內已知動物模型來評價投與免疫原之保護效應。例如,可針對抗體或細胞毒性T細胞之中和能力或免疫T細胞之細胞介素產生能力來分析投與免疫原之個體之免疫血清或免疫T細胞。通常用於此等評估之分析包括(但不限於)病毒中和分析、抗病毒抗原ELISA、干擾素γ細胞介素ELISA、干擾素γ ELISPOT、細胞內多細胞介素染色(ICS)及51鉻釋放細胞毒性分析。亦可使用動物刺激模型來測定免疫原之免疫有效量。 As used herein, "immunologically effective amount" refers to an amount of an immunological response that an immunogen can induce against CMV when administered to a patient, which protects the patient from CMV infection (including primary, recurrent, and/or reinfection) And/or improving at least one condition associated with CMV infection and/or reducing the severity/duration of CMV infection in the patient. This amount should be sufficient to significantly reduce the likelihood or severity of CMV infection. Animal models known in the art can be used to assess the protective effects of the administered immunogen. For example, immune sera or immune T cells of an individual who is administered an immunogen can be analyzed for antibody or cytotoxic T cell neutralizing ability or interleukin production ability of immune T cells. Analysis commonly used for such assessments includes, but is not limited to, virus neutralization assays, antiviral antigen ELISA, interferon gamma interleukin ELISA, interferon gamma ELISPOT, intracellular multicellular staining (ICS), and 51 chrome Release cytotoxicity assay. An animal stimulation model can also be used to determine the immunologically effective amount of the immunogen.

本文所用「條件式複製缺陷型病毒」係指可在某些環境而非其他環境中複製之病毒顆粒。在較佳實施例中,藉由將病毒複製所必需之一或多種蛋白去穩定使病毒成為條件式複製缺陷型病毒。在條件式複製缺陷型病毒中不再存在編碼野生型、非去穩定必需蛋白之核酸。在將一或多種必需蛋白去穩定之條件下,與不具有去穩定必需蛋白之病毒相比,病毒複製減少較佳大於50%、75%、90%、95%、99%或100%。然而,在使去穩定必需蛋白穩定之條件下,病毒複製可在不含有去穩定必需蛋白之CMV之複製量的較佳至少75%、80%、90%、95%、99%或100%下發生。在更佳實施例中,藉由與諸如FKBP或其衍生物等去穩定蛋白 融合來將一或多種必需蛋白去穩定。可藉由諸如Shield-1等穩定劑之存在來穩定此等融合蛋白。本文所用「rdCMV」係指條件式複製缺陷型細胞巨大病毒。 As used herein, "conditional replication-defective virus" refers to viral particles that can be replicated in certain environments, but not in other environments. In a preferred embodiment, the virus is rendered a conditional replication-deficient virus by destabilizing one or more proteins necessary for viral replication. Nucleic acids encoding wild-type, non-destable essential proteins are no longer present in conditional replication-deficient viruses. Under conditions in which one or more essential proteins are destabilized, the reduction in viral replication is preferably greater than 50%, 75%, 90%, 95%, 99%, or 100% compared to a virus that does not have a destabilizing essential protein. However, under conditions which stabilize the destabilizing essential protein, viral replication may be preferably at least 75%, 80%, 90%, 95%, 99% or 100% of the amount of replication of the CMV which does not contain the destabilizing essential protein. occur. In a more preferred embodiment, by destabilizing the protein with, for example, FKBP or a derivative thereof Fusion to destabilize one or more essential proteins. These fusion proteins can be stabilized by the presence of stabilizers such as Shield-1. As used herein, "rdCMV" refers to a conditional replication-defective cell giant virus.

在較佳實施例中,由複製缺陷型病毒誘導之免疫反應與其活病毒對應部分相比在程度及/或幅度上相同或實質上類似。在其他實施例中,藉由電子顯微術分析所達成複製缺陷型病毒之形態與其活病毒對應部分不可區別或實質上類似。 In a preferred embodiment, the immune response induced by the replication-defective virus is identical or substantially similar in degree and/or magnitude to the corresponding portion of the live virus. In other embodiments, the morphology of the replication-defective virus achieved by electron microscopy analysis is indistinguishable or substantially similar to the corresponding portion of the live virus.

本文所用「FKBP」係指SEQ ID NO:11之去穩定蛋白。含有FKBP之融合蛋白係藉由宿主細胞機器降解。本文所用「FKBP衍生物」係指已藉由一或多個胺基酸取代、缺失及/或添加改變之FKBP蛋白或其部分。FKBP衍生物在融合至蛋白時保留FKBP之實質上所有去穩定性質且亦保留FKBP藉由Shield-1穩定之實質上所有能力。較佳FKBP衍生物在所示胺基酸位置處具有以下取代中之一或多者:F15S、V24A、H25R、F36V、E60G、M66T、R71G、D100G、D100N、E102G、K105I及L106P。具有F36V及L106P取代之FKBP衍生物(SEQ ID NO:12)尤佳。在較佳實施例中,編碼FKBP或FKBP衍生物之核酸含有至少一些在人類中並不常用於內源性FKBP之密碼子。此可降低融合蛋白之FKBP或FKBP衍生物與其在人類基因組中之對應部分重排或重組的可能性。核酸序列SEQ ID NO:13使用此等密碼子編碼SEID NO:12。 As used herein, "FKBP" refers to the destabilizing protein of SEQ ID NO:11. Fusion proteins containing FKBP are degraded by host cell machinery. As used herein, "FKBP derivative" refers to an FKBP protein or portion thereof that has been altered, deleted, and/or altered by one or more amino acids. The FKBP derivative retains substantially all of the destabilizing properties of FKBP when fused to the protein and also retains substantially all of the ability of FKBP to be stabilized by Shield-1. Preferably, the FKBP derivative has one or more of the following substitutions at the position of the amino acid shown: F15S, V24A, H25R, F36V, E60G, M66T, R71G, D100G, D100N, E102G, K105I and L106P. A FKBP derivative (SEQ ID NO: 12) having a F36V and L106P substitution is particularly preferred. In a preferred embodiment, the nucleic acid encoding a FKBP or FKBP derivative contains at least some codons that are not commonly used in humans for endogenous FKBP. This may reduce the likelihood that the FKBP or FKBP derivative of the fusion protein will rearrange or recombine with its corresponding portion in the human genome. The nucleic acid sequence SEQ ID NO: 13 encodes SEID NO: 12 using these codons.

本文所用術語「Shield-1」或「Shld 1」係指結合野生型 FKBP及其衍生物且用作穩定劑之合成小分子。與野生型FKBP相比,與F36V衍生物結合緊約1,000倍(Clackson等人,1998,PNAS 95:10437-42)。Shield-1可經合成(基本上如以下文獻中所述:Holt等人,1993,J.Am.Chem.Soc.115:9925-38及Yang等人,2000,J.Med.Chem.43:1135-42及Grimley等人,2008,Bioorganic & Medicinal Chemistry Letters 18:759)或購自Cheminpharma LLC(Farmington,CT)或Clontech Laboratories公司(Mountain View,CA)。亦可在本發明方法中使用Shield-1之鹽。Shield-1具有以下結構: The term "Shield-1" or "Shld 1" as used herein refers to a synthetic small molecule that binds to wild-type FKBP and its derivatives and acts as a stabilizer. The binding to the F36V derivative is approximately 1,000-fold compared to wild-type FKBP (Clackson et al., 1998, PNAS 95: 10437-42). Shield-1 can be synthesized (essentially as described in the following literature: Holt et al, 1993, J. Am. Chem. Soc. 115: 9925-38 and Yang et al, 2000, J. Med. Chem. 43: 1135-42 and Grimley et al., 2008, Bioorganic & Medicinal Chemistry Letters 18: 759) or purchased from Cheminpharma LLC (Farmington, CT) or Clontech Laboratories (Mountain View, CA). Salts of Shield-1 can also be used in the process of the invention. Shield-1 has the following structure:

本文所用術語「融合」或「融合蛋白」係指兩條作為相同鄰接胺基酸序列一部分於框架內佈置之多肽。融合可係直接的,從而使得在多肽間不存在其他胺基酸殘基;或係間接的,從而使得存在小胺基酸連接體,以改良性能或增加功能性。在較佳實施例中,融合係直接的。 The term "fusion" or "fusion protein" as used herein, refers to two polypeptides that are arranged within the framework as part of the same contiguous amino acid sequence. The fusion may be direct such that no additional amino acid residues are present between the polypeptides; or indirectly, such that a small amino acid linker is present to improve performance or increase functionality. In the preferred embodiment, the fusion is straightforward.

本文所用術語「五聚gH複合物」或「gH複合物」係指CMV病毒粒子表面上之五種病毒蛋白之複合物。該複合物係由藉由組裝至gH/gL支架上之UL128、UL130及UL131編碼之蛋白構成(Wang及Shenk,2005 Proc Natl Acad Sci USA.102:1815;Ryckman等人,2008 J.Virol.82:60)。來自CMV株AD 169之複合物蛋白之序列以下列GenBank登錄號顯示:NP_783797.1(UL128)、NP_040067(UL130)、CAA35294.1(UL131)、NP_040009(gH,亦稱為UL75)及NP_783793(gL,亦稱為UL115)。一些經衰減CMV株在UL131中具有一或多種突變,從而使得不表現該蛋白且因此不形成gH複合物。在此等情形下,應修復UL131(使用諸如Wang及Shenk,2005 J.Virol.79:10330中方法等方法),從而使得在本發明rdCMV中表現gH複合物。本發明病毒表現病毒構成五聚gH複合物並在病毒包膜上組裝五聚gH複合物之五種蛋白。 The term "pentameric gH complex" or "gH complex" as used herein refers to a complex of five viral proteins on the surface of CMV virions. The complex consists of proteins encoded by UL128, UL130 and UL131 assembled onto gH/gL scaffolds (Wang and Shenk, 2005 Proc Natl Acad Sci) USA. 102: 1815; Ryckman et al., 2008 J. Virol. 82: 60). The sequence of the complex protein from the CMV strain AD 169 is shown by the following GenBank accession numbers: NP_783797.1 (UL128), NP_040067 (UL130), CAA35294.1 (UL131), NP_040009 (gH, also known as UL75), and NP_783793 (gL) , also known as UL115). Some attenuated CMV strains have one or more mutations in UL131 such that the protein is not expressed and thus does not form a gH complex. In such cases, UL131 (using methods such as those in Wang and Shenk, 2005 J. Virol. 79: 10330) should be repaired such that the gH complex is expressed in the rdCMV of the present invention. The virus of the present invention represents a virus that constitutes a pentameric gH complex and assembles five proteins of a pentameric gH complex on the viral envelope.

本文所用「必需蛋白」係指活體內及組織培養中病毒複製所需之病毒蛋白。CMV中之必需蛋白之實例包括(但不限於)IE1/2、UL37x1、UL44、UL51、UL52、UL53、UL56、UL77、UL79、UL84、UL87及UL105。 As used herein, "essential protein" refers to the viral proteins required for viral replication in vivo and in tissue culture. Examples of essential proteins in CMV include, but are not limited to, IE1/2, UL37x1, UL44, UL51, UL52, UL53, UL56, UL77, UL79, UL84, UL87, and UL105.

本文所用「去穩定必需蛋白」係指經表現並在病毒複製中執行其功能且在不存在穩定劑下降解之必需蛋白。在較佳實施例中,將必需蛋白融合至諸如FKBP或其衍生物等去穩定蛋白。在正常生長條件(即,不存在穩定劑)下,融合蛋白會有所表現,但藉由宿主細胞機器降解。降解不允許必需蛋白在病毒複製中起作用,因此在功能上剔除必需蛋白。在存在諸如Shield-1等穩定劑之條件下,融合蛋白係穩定的且可以維持病毒複製之量執行其功能,該複製係不含有去穩定必需蛋白之CMV之複製量的較佳至少75%、 80%、90%、95%、99%或100%。 As used herein, "de-stabilizing essential protein" refers to an essential protein that is expressed and performs its function in viral replication and is degraded in the absence of a stabilizer. In a preferred embodiment, the essential protein is fused to a destabilizing protein such as FKBP or a derivative thereof. Under normal growth conditions (ie, in the absence of a stabilizer), the fusion protein will behave but degraded by host cell machinery. Degradation does not allow essential proteins to play a role in viral replication, thus functionally eliminating essential proteins. In the presence of a stabilizer such as Shield-1, the fusion protein is stable and can perform its function by maintaining the amount of viral replication which is preferably at least 75% of the amount of replication of the CMV that does not contain the essential protein. 80%, 90%, 95%, 99% or 100%.

複製缺陷型CMVReplication defective CMV

本發明方法使用表現五聚gH複合物之複製缺陷型CMV(rdCMV)。可根據本發明方法使表現五聚gH複合物之任何經衰減CMV病毒成為複製缺陷型。在一個實施例中,經衰減CMV係因修復UL131基因中之突變而已恢復gH複合物表現之AD 169(參見實例1)。 The method of the invention uses a replication defective CMV (rdCMV) that exhibits a pentameric gH complex. Any attenuated CMV virus that exhibits a pentameric gH complex can be made replication defective according to the methods of the invention. In one embodiment, the attenuated CMV has restored AD 169 of gH complex expression by repairing mutations in the UL131 gene (see Example 1).

條件式複製缺陷型病毒係一或多種必需病毒蛋白已藉由必需蛋白之去穩定對應部分替代之突變體。去穩定對應部分係藉由編碼必需蛋白與去穩定蛋白間之融合蛋白之核酸編碼。去穩定必需蛋白僅可在存在穩定劑時起作用以支持病毒複製。在較佳實施例中,使用美國專利公開案2009/0215169中所述方法來賦予表現五聚gH複合物之CMV以條件式複製缺陷型表型。簡言之,將CMV複製所必需之一或多種蛋白融合至去穩定蛋白FKBP或FKBP衍生物。rdCMV中不再存在編碼野生型必需蛋白之核酸。在存在外源添加之可透過細胞之小分子穩定劑Shield-1(Shld-1)下,融合蛋白係穩定的且必需蛋白可起作用以支持病毒複製。rdCMV在存在穩定劑下之複製較佳係不含有去穩定融合蛋白之CMV(例如,用於構築rdCMV之親代經衰減CMV)之複製量的至少75%、80%、90%、95%、99%或100%。在Shield-1不存在下,融合蛋白之去穩定蛋白引導融合蛋白藉由宿主細胞機器實質上降解。在不存在必需蛋白或存在最低量之必需蛋白下,CMV不能以一定量複製以 在患者中產生或維持CMV感染。rdCMV在不存在穩定劑下之複製不會發生或與不含有去穩定融合蛋白之CMV(例如,用於構築rdCMV之親代經衰減CMV)相比減少較佳大於50%、75%、90%、95%或99%。 A conditional replication-deficient virus is one in which one or more essential viral proteins have been stabilized by a corresponding protein to stabilize the corresponding partial replacement. The destabilizing corresponding portion is encoded by a nucleic acid encoding a fusion protein between the essential protein and the destabilizing protein. Destabilizing essential proteins can only function in the presence of stabilizers to support viral replication. In a preferred embodiment, the method described in U.S. Patent Publication No. 2009/0215169 is used to impart a conditional replication defective phenotype to CMVs that exhibit pentameric gH complexes. Briefly, one or more proteins necessary for CMV replication are fused to a destabilizing protein FKBP or FKBP derivative. Nucleic acids encoding wild type essential proteins are no longer present in rdCMV. The fusion protein is stable and the essential protein acts to support viral replication in the presence of an exogenously added cell-permeable small molecule stabilizer Shield-1 (Shld-1). The replication of rdCMV in the presence of a stabilizer is preferably at least 75%, 80%, 90%, 95% of the amount of replication of the CMV that does not contain the destabilizing fusion protein (eg, the parental decaying CMV used to construct rdCMV). 99% or 100%. In the absence of Shield-1, the destabilizing protein of the fusion protein directs the fusion protein to be substantially degraded by the host cell machinery. CMV cannot be replicated in a certain amount in the absence of essential protein or in the presence of a minimum amount of essential protein CMV infection is produced or maintained in the patient. The replication of rdCMV in the absence of a stabilizer does not occur or is preferably greater than 50%, 75%, 90% compared to CMVs that do not contain a destabilizing fusion protein (eg, the parental attenuated CMV used to construct rdCMV). , 95% or 99%.

使用業內所熟知之重組DNA方法,將編碼用於CMV複製及/或建立/維持CMV感染之必需蛋白之核酸附著至編碼FKBP或其衍生物之核酸。經編碼融合蛋白包含在框架內融合至必需蛋白之FKBP或FKBP衍生物。經編碼融合蛋白在Shield-1存在下穩定。然而,經編碼融合蛋白在Shield-1不存在下去穩定且導向破壞。在較佳實施例中,FKBP係SEQ ID NO:11。在其他實施例中,FKBP衍生物係包含一或多個選自由以下之群之胺基酸取代之FKBP:F15S、V24A、H25R、F36V、E60G、M66T、R71G、D100G、D100N、E102G、K105I及L106P。在更佳實施例中,FKBP衍生物包含F36V及/或L106P取代(SEQ ID NO:12)。在更佳實施例中,FKBP衍生物係由SEQ ID NO:13編碼。 A nucleic acid encoding an essential protein for CMV replication and/or establishing/maintaining CMV infection is attached to a nucleic acid encoding FKBP or a derivative thereof using recombinant DNA methods well known in the art. The encoded fusion protein comprises a FKBP or FKBP derivative fused to the essential protein within the framework. The encoded fusion protein is stable in the presence of Shield-1. However, the encoded fusion protein was stable and directed disrupted in the absence of Shield-1. In a preferred embodiment, FKBP is SEQ ID NO:11. In other embodiments, the FKBP derivative comprises one or more FKBPs selected from the group consisting of amino acids: F15S, V24A, H25R, F36V, E60G, M66T, R71G, D100G, D100N, E102G, K105I and L106P. In a more preferred embodiment, the FKBP derivative comprises a F36V and/or L106P substitution (SEQ ID NO: 12). In a more preferred embodiment, the FKBP derivative is encoded by SEQ ID NO:13.

導向藉由與FKBP或其衍生物融合來去穩定之必需蛋白1)係病毒複製所必需;2)可適應去穩定蛋白之融合而不實質上破壞必需蛋白之功能;且3)可適應在編碼必需蛋白之病毒ORF之5'或3'端處插入編碼FKBP或其衍生物之核酸而不實質上破壞其他周圍病毒基因之ORF。在較佳實施例中,導向藉由與FBBP或其衍生物融合來去穩定之必需蛋白編碼非結構蛋白且因此封裝至重組CMV病毒粒子中之可能性降低。表1顯示滿足上述標準之CMV基因。 Orientation of essential proteins destabilized by fusion with FKBP or a derivative thereof 1) is required for viral replication; 2) can be adapted to destabilize protein fusion without substantially disrupting the function of the essential protein; and 3) can be adapted to encode A nucleic acid encoding FKBP or a derivative thereof is inserted at the 5' or 3' end of the viral ORF of the protein without substantially destroying the ORF of other surrounding viral genes. In a preferred embodiment, the possibility that the essential protein destabilized by fusion with FBBP or a derivative thereof encodes a non-structural protein and thus is encapsulated into a recombinant CMV virion is reduced. Table 1 shows the CMV genes that meet the above criteria.

本發明涵蓋包含融合蛋白之rdCMV,該等蛋白具有融合至去穩定蛋白之必需蛋白或其衍生物。必需蛋白衍生物相對於野生型必需蛋白含有一或多個胺基酸取代、添加及/或缺失,但仍可提供必需蛋白至少足以支持在Shield-1存在下之病毒複製之活性。量測病毒活性之實例提供於下文實例中。可使用業內已知方法來測定所關注CMV必需蛋白與衍生物間之差異度。在一個實施例中,使用序列一致性來確定相關性。本發明衍生物將較佳與基本序列至少85%一致、至少90%一致、至少95%一致、至少97%一致、至少99%一致。一致性百分比定義為一致殘基之數目除以殘基總數且乘以100。若比對中之序列具有不同長度(因間隙 或延伸所致),則將在計算中使用代表總長度值之最長序列之長度。 The invention encompasses rdCMV comprising a fusion protein having an essential protein fused to a destabilizing protein or a derivative thereof. The essential protein derivative contains one or more amino acid substitutions, additions and/or deletions relative to the wild type essential protein, but still provides the essential protein at least sufficient to support viral replication in the presence of Shield-1. Examples of measuring viral activity are provided in the examples below. Differences between the essential proteins and derivatives of the CMV of interest can be determined using methods known in the art. In one embodiment, sequence consistency is used to determine correlation. The derivatives of the invention will preferably be at least 85% identical, at least 90% identical, at least 95% identical, at least 97% identical, at least 99% identical to the base sequence. The percent identity is defined as the number of consistent residues divided by the total number of residues and multiplied by 100. If the sequences in the alignment have different lengths (due to the gap) Or the extension, the length of the longest sequence representing the total length value will be used in the calculation.

在一些實施例中,病毒複製所必需之一或多種導向去穩定之病毒蛋白係選自由以下組成之群:IE1/2、UL51、UL52、UL84、UL79、UL87、UL37x1、UL77及UL53或其衍生物。在具體實施例中,病毒複製所必需之一或多種導向去穩定之病毒蛋白係選自由以下組成之群:IE1/2、UL51、UL52、UL84、UL79、UL87。在更具體實施例中,病毒複製所必需之一或多種導向去穩定之病毒蛋白係選自由以下組成之群:IE1/2、UL51、UL52、UL79及UL84。 In some embodiments, one or more of the directed destabilizing viral proteins necessary for viral replication are selected from the group consisting of: IE1/2, UL51, UL52, UL84, UL79, UL87, UL37x1, UL77, and UL53 or derivatives thereof Things. In a particular embodiment, one or more of the directed destabilizing viral proteins necessary for viral replication are selected from the group consisting of IE1/2, UL51, UL52, UL84, UL79, UL87. In a more specific embodiment, one or more of the directed destabilizing viral proteins necessary for viral replication are selected from the group consisting of IE1/2, UL51, UL52, UL79, and UL84.

一種以上必需蛋白可藉由與FKBP或其衍生物融合去穩定。在一些實施例中,該等必需蛋白在CMV複製及/或感染之不同階段(包括,但不限於,立即早期、早期或晚期)作用。在較佳實施例中,病毒複製所必需之經導向去穩定之病毒蛋白的組合係選自由以下組成之群:IE1/2與UL51、IE1/2與UL52、IE1/2與UL79、IE1/2與UL84、UL84與UL51,及UL84與UL52。在一個更佳實施例中,IE1/2與UL51係於相同重組CMV中導向去穩定。在一個最佳實施例中,包含IE1/2之融合蛋白係SEQ ID NO:1且包含UL51之融合蛋白係SEQ ID NO:3。SEQ ID NO:1及3可分別由SEQ ID No:2及4編碼。具有去穩定之IE 1/2及UL51之rdCMV之基因組示於SEQ ID NO:14中。 More than one essential protein can be destabilized by fusion with FKBP or a derivative thereof. In some embodiments, the essential proteins act at different stages of CMV replication and/or infection, including, but not limited to, immediate early, early, or late. In a preferred embodiment, the combination of directed destabilizing viral proteins necessary for viral replication is selected from the group consisting of IE1/2 and UL51, IE1/2 and UL52, IE1/2 and UL79, IE1/2 With UL84, UL84 and UL51, and UL84 and UL52. In a more preferred embodiment, IE1/2 and UL51 are oriented destabilized in the same recombinant CMV. In a preferred embodiment, the fusion protein comprising IE1/2 is SEQ ID NO: 1 and the fusion protein comprising UL51 is SEQ ID NO: 3. SEQ ID NOS: 1 and 3 can be encoded by SEQ ID Nos: 2 and 4, respectively. The genome of rdCMV with destabilized IE 1/2 and UL51 is shown in SEQ ID NO: 14.

FKBP或其衍生物可直接地或間接地融合至必需蛋白。 在較佳實施例中,FKBP或其衍生物係直接地融合至必需蛋白。 FKBP or a derivative thereof can be directly or indirectly fused to an essential protein. In a preferred embodiment, FKBP or a derivative thereof is directly fused to an essential protein.

FKBP或其衍生物可在必需蛋白之N或C端融合至必需蛋白。在較佳實施例中,FKBP係融合至必需蛋白之N端。 FKBP or a derivative thereof can be fused to an essential protein at the N or C terminus of an essential protein. In a preferred embodiment, the FKBP line is fused to the N-terminus of the essential protein.

一種以上FKBP或其衍生物可融合至必需蛋白。在一種以上FKBP或其衍生物融合至必需蛋白之實施例中,各個FKBP或其衍生物可相同或不同。在較佳實施例中,一種FKBP或其衍生物融合至必需蛋白。 More than one FKBP or a derivative thereof can be fused to an essential protein. In embodiments in which more than one FKBP or derivative thereof is fused to an essential protein, each FKBP or derivative thereof may be the same or different. In a preferred embodiment, a FKBP or derivative thereof is fused to an essential protein.

其他不活化方法Other inactivation methods

在一些實施例中,進一步使用化學或物理不活化使上文所述rdCMV不活化。此等方法之實例包括熱處理,與甲醛、β-丙內酯(BPL)或二乙烯亞胺(BEI)一起培育,或γ輻照。較佳方法不破壞或不實質上破壞免疫原性,包括(但不限於)由五聚gH複合物誘導之免疫原性。由此,與無其他不活化處理之rdCMV相比,由已進一步不活化之CMV誘發之免疫反應保持或實質上保持。在較佳實施例中,進一步不活化之CMV誘導中和抗體之能力與彼等藉由無其他不活化處理治療之rdCMV誘導者相當。以經驗決確定藉由化學或物理方法中之任一者或組合之不活化方案以確保CMV(包括五聚gH複合物)在內之CMV之免疫原性。 In some embodiments, the rdCMV described above is further inactivated using chemical or physical inactivation. Examples of such methods include heat treatment, incubation with formaldehyde, beta-propiolactone (BPL) or diethyleneimine (BEI), or gamma irradiation. Preferred methods do not destroy or substantially destroy immunogenicity, including, but not limited to, immunogenicity induced by the pentameric gH complex. Thus, the CMV-induced immune response is maintained or substantially maintained as compared to rdCMV without other inactivation treatment. In a preferred embodiment, the ability of the further inactivated CMV to induce neutralizing antibodies is comparable to those of the rdCMV inducers treated by no other inactivation treatment. The immunogenicity of CMV, including CMV (including pentameric gH complex), is determined empirically by an inactivation scheme by either or both of chemical or physical methods.

病毒複製之評估Evaluation of viral replication

熟習此項技術者可使用病毒複製分析來確定融合至FKBP或其衍生物之特定必需蛋白之實用性。由於在Shield-1存在下FKBP或其衍生物與必需蛋白之附著不應實 質上影響基因表現/經編碼產物功能,因此rdCMV應以在Shield-1存在下與親代CMV相當之比率(較佳係親代病毒量之至少75%、80%、90%、95%、99%或100%)複製。在不存在Shieid-1下,rdCMV之複製與親代CMV實質上不同(與不含有去穩定融合蛋白之CMV相比減少較佳大於50%、75%、90%、95%、99%或100%)。 Those skilled in the art can use viral replication assays to determine the utility of a particular essential protein fused to FKBP or a derivative thereof. Due to the attachment of FKBP or its derivatives to essential proteins in the presence of Shield-1 Qualitatively affects gene expression/coding product function, therefore rdCMV should be in a ratio comparable to parental CMV in the presence of Shield-1 (preferably at least 75%, 80%, 90%, 95% of the amount of parental virus, 99% or 100%) copy. In the absence of Shieid-1, replication of rdCMV is substantially different from that of parental CMV (reduced by more than 50%, 75%, 90%, 95%, 99% or 100 compared to CMV without destabilizing fusion protein) %).

在較佳實施例中,在存在至少2 μM Shield-1下,rdCMV複製非rdCMV複製量之較佳至少90%、更佳至少95%、最佳至少99%。 In a preferred embodiment, the rdCMV replicates preferably at least 90%, more preferably at least 95%, and most preferably at least 99% of the non-rdCMV replication in the presence of at least 2 μM Shield-1.

在一個實施例中,在存在至少2 μM Shield-1下,包含本發明rdCMV之組合物之病毒效價係至少105 pfu/ml、更佳至少107 pfu/ml。 In one embodiment, the viral titer comprising the composition of the rdCMV of the invention is at least 10 5 pfu/ml, more preferably at least 10 7 pfu/ml, in the presence of at least 2 μM Shield-1.

反之,在Shield-1不存在下,rdCMV實質上不應複製。複製缺陷型機制之品質係藉由在不允許病毒複製之條件下控制之嚴格程度(即,子代病毒粒子在該等條件下之感染效價)來判斷。在Shield-1不存在下,本發明rdCMV實質上不能複製(在細胞培養物中或在患者內)。其在ARPE-19細胞及其他類型人類原代細胞中之複製係有條件的,且需要Shield-1在培養基中之莫耳濃度大於0.1 μM、較佳至少2 μM以維持病毒複製。 Conversely, in the absence of Shield-1, rdCMV should not be replicated in nature. The quality of the replication defective mechanism is judged by the degree of rigor of control under conditions that do not allow for viral replication (i.e., the infectious virion of the progeny virions under such conditions). In the absence of Shield-1, the rdCMV of the invention is substantially incapable of replication (in cell culture or within a patient). Its replication in ARPE-19 cells and other types of human primary cells is conditional and requires a molar concentration of Shield-1 in the culture medium greater than 0.1 μM, preferably at least 2 μM to maintain viral replication.

在一個實施例中,在Shield-1不存在下,包含本發明rdCMV之組合物之病毒效價小於2 pfu/ml、更佳小於1 pfu/ml。 In one embodiment, the composition comprising the rdCMV of the invention has a virus titer of less than 2 pfu/ml, more preferably less than 1 pfu/ml, in the absence of Shield-1.

可使用評價CMV複製之方法來評價在Shield-1不存在或 存在下之rdCMV複製。然而,在較佳實施例中,使用TCID50。 The method of evaluating CMV replication can be used to evaluate the absence of Shield-1 or There is rdCMV replication underneath. However, in the preferred embodiment, TCID 50 is used.

在另一實施例中,rdCMV效價係藉由50%組織培養感染劑量(TCID50)分析測定。簡言之,此稀釋分析量化殺傷50%受感染宿主所需病毒之量。平鋪宿主細胞(例如,ARPE-19細胞)且添加病毒之連續稀釋物。在培育後,觀察並記錄各病毒稀釋度之細胞死亡(即受感染細胞)之百分比。使用結果來以數學方式計算TCID50。 In another embodiment, the rdCMV titer is determined by a 50% tissue culture infectious dose (TCID50) assay. Briefly, this dilution analysis quantifies the amount of virus required to kill 50% of infected hosts. Host cells (eg, ARPE-19 cells) are plated and serial dilutions of the virus are added. After incubation, the percentage of cell death (i.e., infected cells) for each virus dilution was observed and recorded. Use the results to mathematically calculate TCID50.

在另一實施例中,rdCMV效價係使用噬菌斑分析測定。病毒噬菌斑分析測定病毒試樣中噬菌斑形成單位(pfu)之數目。簡言之,宿主細胞(例如,ARPE-19細胞)之鋪滿單層經不同稀釋度之rdCMV感染且經半固體培養基(例如瓊脂或羧甲基纖維素)覆蓋,以防止病毒感染任意地傳播。病毒噬菌斑係在病毒感染固定細胞單層內之細胞時形成。病毒感染細胞將溶解並將感染傳播至毗鄰細胞,其中重複感染-溶解循環。受感染細胞區域將產生噬菌斑(由未受感染細胞包圍之感染區域),其可目視或利用光學顯微鏡觀察。對噬菌斑計數且使用與用於準備板之稀釋因子組合之結果來計算噬菌斑形成單位/試樣單位體積(pfu/mL)之數目。pfu/mL結果代表試樣內感染顆粒之數目且係基於所形成各噬菌斑皆代表一個感染病毒顆粒之假定。 In another embodiment, the rdCMV titer is determined using plaque assay. Viral plaque assay determines the number of plaque forming units (pfu) in the virus sample. Briefly, a confluent monolayer of host cells (eg, ARPE-19 cells) is infected with different dilutions of rdCMV and covered with semi-solid medium (eg, agar or carboxymethylcellulose) to prevent viral infection from spreading arbitrarily . Viral plaques are formed when the virus infects cells in a fixed cell monolayer. The virus-infected cells will dissolve and spread the infection to adjacent cells where the infection-dissolution cycle is repeated. The infected cell area will produce plaques (infected areas surrounded by uninfected cells) which can be visually observed or observed using an optical microscope. The number of plaque forming units per sample unit volume (pfu/mL) was calculated by counting the plaques and using the results in combination with the dilution factors used to prepare the plates. The pfu/mL results represent the number of infectious particles in the sample and are based on the assumption that each plaque formed represents an infected virus particle.

在另一實施例中,使用hu-SCID小鼠模型來評估rdCMV在活體內複製之能力。簡言之,以外科手術方式將數片人類胎兒組織(例如胸腺及肝)植入SCID小鼠之腎囊膜中。在 2至3個月後,當人類組織血管化時,接種rdCMV。在接種後3至4週在噬菌斑分析中評價病毒效價。可在Shield-1不存在或存在下執行動物實驗。 In another embodiment, the hu-SCID mouse model is used to assess the ability of rdCMV to replicate in vivo. Briefly, several human fetal tissues, such as the thymus and liver, are surgically implanted into the renal capsule of SCID mice. in After 2 to 3 months, when human tissue is vascularized, rdCMV is inoculated. Viral titers were evaluated in plaque assays 3 to 4 weeks after inoculation. Animal experiments can be performed in the absence or presence of Shield-1.

免疫反應之評估Assessment of immune response

將本發明rdCMV投與患者誘發對CMV之免疫反應、較佳保護性免疫反應,該反應可治療患者之CMV感染或與此一感染相關之病狀及/或降低其可能性。免疫反應至少部分地係針對五聚gH複合物。 Administration of the rdCMV of the invention induces an immune response to CMV, preferably a protective immune response, which can treat a patient's CMV infection or a condition associated with the infection and/or reduce its likelihood. The immune response is at least partially directed to the pentameric gH complex.

可使用業內已知方法來評價藉由rdCMV誘發之免疫反應。 The immune response induced by rdCMV can be assessed using methods known in the art.

可使用業內已知動物模型來評價投與rdCMV之保護效應。在一個實施例中,可分析投與rdCMV之個體之免疫血清之中和能力,包括但不限於對病毒附著或進入宿主細胞之阻斷。在其他實施例中,可分析投與rdCMV之個體之T細胞在存在所關注抗原下之細胞介素產生能力,包括(但不限於)干擾素γ。亦可使用動物刺激模型來測定免疫原之免疫有效量。 Animal models known in the art can be used to evaluate the protective effects of administration of rdCMV. In one embodiment, the immune sera neutralization ability of an individual administered rdCMV can be analyzed, including but not limited to blocking of the virus or entry into the host cell. In other embodiments, the T cells of an individual administered with rdCMV can be analyzed for the ability to produce interleukins in the presence of the antigen of interest, including but not limited to, interferon gamma. An animal stimulation model can also be used to determine the immunologically effective amount of the immunogen.

病毒中和係指病毒特異性抗體能中斷培養物中之病毒進入及/或複製。量測中和活性之常見分析係病毒噬菌斑減少分析。本發明中之中和分析係指可阻斷病毒進入細胞之血清滴定。NT50效價定義為在病毒中和分析中阻斷50%輸入病毒之血清稀釋度的倒數。自非線性邏輯四參數曲線擬合獲得NT50效價。 Viral neutralization refers to the ability of a virus-specific antibody to disrupt viral entry and/or replication in culture. A common assay for measuring neutralizing activity is viral plaque reduction assay. Neutralization assays in the present invention refer to serum titration that blocks the entry of a virus into a cell. The NT50 titer is defined as the reciprocal of the serum dilution that blocks 50% of the input virus in the virus neutralization assay. The NT50 titer was obtained from a nonlinear logic four-parameter curve fit.

複製缺陷型CMV之製造Manufacture of replication defective CMV

本發明涵蓋製備rdCMV之方法。在存在諸如Shield-1等穩定劑下在上皮細胞上、較佳人類上皮細胞且更佳人類視網膜色素上皮細胞上繁殖本發明rdCMV。在其他實施例中,人類視網膜色素上皮細胞係以登錄號CRL-2302寄存於美國典型培養物保藏中心(American Type Culture Collection,ATCC)之ARPE-19細胞。在一些實施例中,Shield-1係以至少0.5 μM之濃度存在於組織培養基中。在較佳實施例中,Shield-1係以至少2.0 μM之濃度存在於組織培養基中。 The invention encompasses methods of making rdCMV. The rdCMV of the present invention is propagated on epithelial cells, preferably human epithelial cells, and more preferably human retinal pigment epithelial cells in the presence of a stabilizer such as Shield-1. In other embodiments, the human retinal pigment epithelial cell line is deposited with ARPE-19 cells of the American Type Culture Collection (ATCC) under accession number CRL-2302. In some embodiments, the Shield-1 is present in the tissue culture medium at a concentration of at least 0.5 μM. In a preferred embodiment, the Shield-1 is present in the tissue culture medium at a concentration of at least 2.0 μM.

在一些實施例中,用於繁殖rdCMV之細胞係在微載體上生長。微載體係允許附著細胞在旋轉燒瓶或生物反應器(例如旋轉壁微重力生物反應器及流化床生物反應器)中生長之支持基質。微載體通常係125-250 μM球體,其密度允許其在輕緩攪拌的同時維持在懸浮液中。微載體可自諸多不同材料製得,包括(但不限於)DEAE-葡聚糖、玻璃、聚苯乙烯塑膠、丙烯醯胺及膠原。微載體可具有不同表面成份,包括(但不限於)細胞外基質蛋白、重組蛋白、肽及帶電分子。亦可使用其他高密度細胞培養系統,例如Corning HyperFlask®及HyperStack®系統。 In some embodiments, the cell line used to propagate rdCMV is grown on a microcarrier. Microcarriers are support matrices that allow adherent cells to grow in a rotating flask or bioreactor, such as a rotating wall microgravity bioreactor and a fluidized bed bioreactor. Microcarriers are typically 125-250 μM spheres, the density of which allows them to be maintained in suspension while gently agitating. Microcarriers can be made from a variety of different materials including, but not limited to, DEAE-dextran, glass, polystyrene plastic, acrylamide, and collagen. Microcarriers can have different surface components including, but not limited to, extracellular matrix proteins, recombinant proteins, peptides, and charged molecules. Other high-density cell culture systems such as Corning HyperFlask® and HyperStack® systems can also be used.

可採集無細胞組織培養基且可自其純化rdCMV。CMV病毒顆粒之直徑係約200 nm且可使用業內已知技術將其與自存於經收穫培養基中之其他蛋白分離,該等技術包括(但不限於)經由密度梯度或20%山梨醇墊(Sorbitol cushion)之超速離心。疫苗之蛋白質量可藉由Bradford分析測定。 Cell-free tissue culture medium can be harvested and rdCMV can be purified therefrom. The CMV virions are about 200 nm in diameter and can be separated from other proteins present in the harvested medium using techniques known in the art including, but not limited to, via a density gradient or a 20% sorbitol mat ( Ultracentrifugation of Sorbitol cushion). The amount of protein in the vaccine can be determined by Bradford analysis.

可使用Shield-1結合FKBP來控制rdCMV之複製。在完成 組織培養細胞中期望量之病毒繁殖後,複製能力不再合意。自rdCMV抽出Shield-1以使病毒複製為缺陷型(例如,以投與患者)。在一個實施例中,藉由洗滌一或多次自Shield-1純化rdCMV。在另一實施例中,經由超速離心自Shield-1純化rdCMV。在另一實施例中,經由透析過濾自Shield-1純化rdCMV。透析過濾通常用於純化病毒顆粒。在一個實施例中,使用孔徑為約750千道爾頓(kilodalton)之過濾器,該孔徑將僅允許Shield-1經過孔。 Shield-1 can be used in conjunction with FKBP to control the replication of rdCMV. On completion After the desired amount of virus in the tissue culture cells is propagated, the ability to replicate is no longer satisfactory. Shield-1 is withdrawn from rdCMV to replicate the virus as defective (eg, to the patient). In one embodiment, rdCMV is purified from Shield-1 by washing one or more times. In another embodiment, rdCMV is purified from Shield-1 via ultracentrifugation. In another embodiment, rdCMV is purified from Shield-1 via diafiltration. Diafiltration is typically used to purify viral particles. In one embodiment, a filter having a pore size of about 750 kilodaltons is used, which will only allow Shield-1 to pass through the aperture.

在自Shield-1純化rdCMV後,可存在少量殘留於rdCMV組合物中之殘餘Shield-1。在一個實施例中,在純化後,CMV組合物中之Shld-1之量係維持在組織培養物中之複製所需量之至多1/100。在另一實施例中,在純化後,rdCMV組合物中之Shield-1之量係0.1 μM或更小。在另一實施例中,在純化後,rdCMV組合物中之Shield-1之量不可檢測。 After purification of rdCMV from Shield-1, a small amount of residual Shield-1 remaining in the rdCMV composition may be present. In one embodiment, the amount of Shld-1 in the CMV composition after purification is maintained at up to 1/100 of the amount required for replication in tissue culture. In another embodiment, the amount of Shield-1 in the rdCMV composition is 0.1 μM or less after purification. In another embodiment, the amount of Shield-1 in the rdCMV composition is undetectable after purification.

組合物中之Shield-1量之測定可使用LC/MS(液相層析-質譜)或HPLC/MS(高效液相層析-質譜)分析來檢測。該等技術組合LC或HPLC之物理分離能力與質量分析能力且可檢測複合物混合物中所關注化學品。 The determination of the amount of Shield-1 in the composition can be detected using LC/MS (liquid chromatography-mass spectrometry) or HPLC/MS (high performance liquid chromatography-mass spectrometry) analysis. These techniques combine the physical separation capabilities and mass analysis capabilities of LC or HPLC and detect the chemicals of interest in the composite mixture.

佐劑Adjuvant

佐劑係可輔助免疫原產生免疫反應之物質。佐劑可藉由諸如以下中之一或多者等不同機制起作用:延長抗原生物學或免疫學半衰期;改良至抗原呈遞細胞之抗原遞送;改良藉由抗原呈遞細胞之抗原處理及呈遞;達成劑量節約及 誘導產生免疫調節細胞介素(Vogel,2000,Clin Infect Dis 30:S266)。在一些實施例中,本發明組合物包含rdCMV及佐劑。 An adjuvant is a substance that aids in the immune response of an immunogen. The adjuvant may act by different mechanisms such as one or more of: extending antigenic biology or immunological half-life; improving antigen delivery to antigen presenting cells; improving antigen processing and presentation by antigen presenting cells; Dose savings and An immunomodulatory interleukin is induced (Vogel, 2000, Clin Infect Dis 30: S266). In some embodiments, the compositions of the invention comprise rdCMV and an adjuvant.

多種不同類型之佐劑可用於輔助產生免疫反應。特定佐劑之實例包括氫氧化鋁;磷酸鋁、羥基磷酸鋁、非晶形鋁的羥基磷酸硫酸鹽佐劑(AAHSA)或其他鋁鹽;磷酸鈣;DNA CpG基序;單磷醯脂質A;霍亂毒素;大腸桿菌(E.coli)熱不穩定毒素;百日咳毒素;胞壁醯二肽;弗氏不完全佐劑(Freund's incomplete adjuvant);MF59;SAF;免疫刺激複合物;脂質體;生物可降解微球體;皂苷;非離子阻斷共聚物;胞壁醯肽類似物;聚膦腈;合成聚核苷酸;IFN-γ;IL-2;IL-12;及ISCOMS。(Vogel,2000,Clin Infect Dis 30:S266;Klein等人,2000,J Pharm Sci 89:311;Rimmelzwaan等人,2001,Vaccine 19:1180;Kersten,2003,Vaccine 21:915;O'Hagen,2001,Curr.Drug Target Infect.Disord.1:273)。 A variety of different types of adjuvants can be used to aid in the production of an immune response. Examples of specific adjuvants include aluminum hydroxide; aluminum phosphate, aluminum hydroxyphosphate, amorphous aluminum hydroxyphosphate sulfate adjuvant (AAHSA) or other aluminum salts; calcium phosphate; DNA CpG motif; monophosphonium lipid A; cholera Toxin; E. coli heat labile toxin; pertussis toxin; cell wall 醯 dipeptide; Freund's incomplete adjuvant; MF59; SAF; immunostimulating complex; liposome; biodegradable Microspheres; saponins; non-ionic blocking copolymers; cell wall purine peptide analogs; polyphosphazenes; synthetic polynucleotides; IFN-γ; IL-2; IL-12; (Vogel, 2000, Clin Infect Dis 30: S266; Klein et al, 2000, J Pharm Sci 89: 311; Rimmelzwaan et al, 2001, Vaccine 19: 1180; Kersten, 2003, Vaccine 21: 915; O'Hagen, 2001 , Curr.Drug Target Infect.Disord.1:273).

在一些實施例中,在本發明組合物中使用基於油之佐劑,包括(但不限於)不完全弗氏佐劑及MF59。 In some embodiments, oil-based adjuvants are used in the compositions of the invention, including but not limited to incomplete Freund's adjuvant and MF59.

在其他實施例中,在本發明組合物中使用微粒佐劑,包括(但不限於)ISCOMATRIX®佐劑及/或磷酸鋁佐劑。 In other embodiments, microparticle adjuvants are used in the compositions of the invention, including but not limited to, ISCOMATRIX® adjuvants and/or aluminum phosphate adjuvants.

醫藥組合物Pharmaceutical composition

本發明之另一特徵係本文所述重組CMV在組合物、較佳免疫原性組合物或疫苗中之用途,該組合物或疫苗用於治療具有CMV感染之患者及/或降低CMV感染之可能性。適 宜地,該組合物包含醫藥上可接受之載劑。 Another feature of the invention is the use of a recombinant CMV as described herein in a composition, preferably an immunogenic composition or vaccine, for use in treating a patient having a CMV infection and/or reducing the likelihood of CMV infection Sex. suitable Preferably, the composition comprises a pharmaceutically acceptable carrier.

「醫藥上可接受之載劑」意指可安全地用於全身投與之液體填充劑、稀釋劑或囊封物質。端視特定投與途徑而定,可使用多種業內熟知之醫藥上可接受之載劑。該等載劑可係選自包括以下之群:糖、澱粉、纖維素及其衍生物、麥芽、明膠、滑石粉、硫酸鈣、植物油、合成油、多元醇、海藻酸、磷酸鹽緩衝溶液(包括磷酸鹽緩衝鹽水)、乳化劑、等滲鹽水及無致熱原水。特定而言,醫藥上可接受之載劑可含有不同組份,例如緩衝液、注射用無菌水、生理鹽水或磷酸鹽緩衝鹽水、蔗糖、組胺酸、鹽及聚山梨醇酯。諸如「生理上可接受之」「稀釋劑」或「賦形劑」等術語可互換使用。 "Pharmaceutically acceptable carrier" means a liquid filler, diluent or encapsulating material that can be safely administered for systemic administration. Depending on the particular route of administration, a variety of pharmaceutically acceptable carriers well known in the art can be used. The carriers may be selected from the group consisting of sugars, starches, celluloses and derivatives thereof, malt, gelatin, talc, calcium sulfate, vegetable oils, synthetic oils, polyols, alginic acid, phosphate buffer solutions. (including phosphate buffered saline), emulsifier, isotonic saline and non-pyrogenic water. In particular, pharmaceutically acceptable carriers may contain different components such as buffers, sterile water for injection, physiological saline or phosphate buffered saline, sucrose, histidine, salts and polysorbates. Terms such as "physiologically acceptable", "diluent" or "excipient" are used interchangeably.

疫苗調配用程序揭示於(例如)New Generation Vaccines (1997,Levine等人,Marcel Dekker公司,New York,Basel,Hong Kong)中,其以引用方式併入本文中。 Vaccine formulation procedures are disclosed, for example, in New Generation Vaccines (1997, Levine et al, Marcel Dekker, Inc., New York, Basel, Hong Kong), which is incorporated herein by reference.

調配物Formulation

在一些實施例中,將本發明rdCMV投與患者以誘發免疫反應。期望在免疫原性組合物儲存期間使rdCMV組合物效能之損失最小化或避免該損失。支持此一目的之條件包括(但不限於):(1)持續儲存穩定性,(2)耐受脅迫冷凍-解凍循環,(3)在環境溫度下穩定長達一週,(4)維持免疫原性,(5)與佐劑策略相容。影響rdCMV穩定性之條件包括(但不限於)緩衝液pH、緩衝液離子強度、特定賦形劑之存在/不存在及溫度。組合物包含緩衝液以增加適宜作為疫 苗組合物之經純化rdCMV病毒顆粒之穩定性。 In some embodiments, a rdCMV of the invention is administered to a patient to elicit an immune response. It is desirable to minimize or avoid loss of rdCMV composition efficacy during storage of the immunogenic composition. Conditions that support this purpose include (but are not limited to): (1) sustained storage stability, (2) tolerance to the freeze-thaw cycle, (3) stability at ambient temperature for up to one week, and (4) maintenance of the immunogen Sex, (5) compatible with adjuvant strategies. Conditions that affect the stability of rdCMV include, but are not limited to, buffer pH, buffer ionic strength, presence/absence of specific excipients, and temperature. The composition contains a buffer to increase the suitability as an epidemic The stability of the purified rdCMV virus particles of the seedling composition.

可在小鼠及/或病毒進入分析中藉由免疫原性分析來評估病毒顆粒完整性之保持。病毒進入事件取決於包括五聚gH複合物在內之病毒糖蛋白之完整性及功能。五聚gH複合物亦提供rdCMV之實質性免疫原性,由此將兩種性質關聯。 The maintenance of viral particle integrity can be assessed by immunogenicity analysis in mouse and/or virus entry assays. The viral entry event depends on the integrity and function of the viral glycoprotein including the pentameric gH complex. The pentameric gH complex also provides substantial immunogenicity of rdCMV, thereby correlating the two properties.

在一些實施例中,將rdCMV儲存在包含15-35 mM組胺酸及100-200 mM NaCl之緩衝液(pH介於5與7之間)中。在更具體實施例中,緩衝液包含25 mM組胺酸及150 mM NaCl(pH 6)。 In some embodiments, rdCMV is stored in a buffer (pH between 5 and 7) comprising 15-35 mM histidine and 100-200 mM NaCl. In a more specific embodiment, the buffer comprises 25 mM histidine and 150 mM NaCl (pH 6).

在其他實施例中,可添加糖以提供進一步穩定性,例如多元醇(包括(但不限於)甘露醇及山梨醇);單醇(包括(但不限於)葡萄糖、甘露糖、半乳糖及果糖);二醇(包括(但不限於)乳糖、麥芽糖、蔗糖、乳果糖及海藻糖)及三糖(包括(但不限於)棉子糖及松三糖)。在更具體實施例中,糖係蔗糖。在甚至更具體實施例中,蔗糖介於5%與15%之間。 In other embodiments, sugars may be added to provide further stability, such as polyols (including but not limited to mannitol and sorbitol); monoalcohols including, but not limited to, glucose, mannose, galactose, and fructose Glycols (including but not limited to lactose, maltose, sucrose, lactulose and trehalose) and trisaccharides (including but not limited to, raffinose and melezitose). In a more specific embodiment, the sugar is sucrose. In an even more specific embodiment, the sucrose is between 5% and 15%.

在較佳實施例中,將rdCMV儲存在包含25 mM組胺酸、150 mM NaCl、9%蔗糖(pH 6)之緩衝液中。 In a preferred embodiment, rdCMV is stored in a buffer comprising 25 mM histidine, 150 mM NaCl, 9% sucrose (pH 6).

投與Cast

可使用本文所提供指導連同業內熟知技術將本文所述rdCMV調配並投與患者。醫藥投與用導則通常提供於(例如)以下文獻中:Vaccines,編輯Plotkin及Orenstein,W.B.Sanders公司,1999;Remington's Pharmaceutical Sciences第20版,編輯Gennaro,Mack Publishing,2000;及 Modem Pharmaceutics第2版,編輯Banker及Rhodes,Marcel Dekker公司,1990。 The rdCMV described herein can be formulated and administered to a patient using the guidance provided herein in conjunction with well known techniques in the art. Guidance for the administration of pharmaceuticals is generally provided, for example, in the following literature: Vaccines , ed. Plotkin and Orenstein, WBSanders, 1999; Remington's Pharmaceutical Sciences 20th edition, edited by Gennaro, Mack Publishing, 2000; and Modem Pharmaceutics 2nd edition, Edit Banker and Rhodes, Marcel Dekker, 1990.

疫苗可藉由諸如以下等不同途徑投與:皮下、肌內、靜脈內、黏膜、非經腸、經皮或真皮內。皮下及肌內投與可使用(例如)針頭或噴射注射器執行。在一實施例中,本發明疫苗係經肌內投與。經皮或真皮內遞送可經由真皮內注射器針頭注射或使能器件(例如微米針頭或微米陣列貼片)達成。 Vaccines can be administered by different routes such as subcutaneous, intramuscular, intravenous, mucosal, parenteral, transdermal or intradermal. Subcutaneous and intramuscular administration can be performed using, for example, a needle or a jet injector. In one embodiment, the vaccine of the invention is administered intramuscularly. Transdermal or intradermal delivery can be achieved via intradermal syringe needle injection or enabling devices such as microneedle or microarray patches.

本文所述組合物可以與劑量調配物相容之方式且以可在免疫原性上有效治療CMV感染(包括原發性、復發性及/或再)及/或降低其可能性之量投與。在本發明上下文中,投與患者之劑量應足以隨時間在患者中實現有益反應,例如降低CMV感染程度、改善與CMV感染相關之疾病症狀及/或縮短CMV感染之時長及/或嚴重程度或降低CMV感染(包括原發性、復發性及/或再)之可能性。 The compositions described herein can be administered in a manner compatible with the dosage formulation and in an amount effective to treat CMV infection (including primary, recurrent, and/or re-) and/or reduce the likelihood of immunogenicity. . In the context of the present invention, the dosage administered to the patient should be sufficient to effect a beneficial response in the patient over time, such as reducing the extent of CMV infection, improving the symptoms of the disease associated with CMV infection and/or shortening the duration and/or severity of CMV infection. Or reduce the likelihood of CMV infection (including primary, recurrent, and/or re-).

適宜給藥方案可由彼等熟習此項技術者容易地測定且較佳考慮業內熟知因素測定,該等因素包括患者之年齡、重量、性別及醫學病況;投與途徑;期望效應;及所用特定組合物。在測定欲在針對CMV之治療或預防中投與之rdCMV之有效量時,醫師可評估病毒之循環血漿濃度、疾病進展及/或抗CMV抗體之產生。疫苗組合物之劑量係由103至1012噬菌斑形成單位(pfu)之範圍組成。在不同實施例中,劑量範圍係104至1010 pfu、105至109 pfu、106至108 pfu或該等所述範圍內之任一劑量。當欲投與一種以上疫苗 (即,呈組合疫苗)時,各疫苗劑之量係在其所述範圍內。 Suitable dosing regimens can be readily determined by those skilled in the art and are preferably determined by well-known factors in the industry, including the age, weight, sex, and medical condition of the patient; the route of administration; the desired effect; and the particular combination employed. Things. In determining an effective amount of rdCMV to be administered in the treatment or prevention against CMV, the physician can assess circulating plasma concentrations of the virus, disease progression, and/or production of anti-CMV antibodies. The dosage of the vaccine composition consists of a range of 10 3 to 10 12 plaque forming units (pfu). In various embodiments, the dosage range is from 10 4 to 10 10 pfu, from 10 5 to 10 9 pfu, from 10 6 to 10 8 pfu, or any of these ranges. When more than one vaccine (i.e., in combination vaccine) is to be administered, the amount of each vaccine agent is within its stated range.

疫苗組合物可以單一劑量或多劑量模式投與。疫苗可在投與、鑑別穩定緩衝液及適宜佐劑組合物前數小時或數天利用佐劑製備。疫苗可以通常實踐之體積(在0.1 mL至0.5 mL範圍內)投與。 The vaccine composition can be administered in a single dose or multiple dose mode. The vaccine can be prepared with an adjuvant several hours or days prior to administration, identification of a stabilizing buffer, and a suitable adjuvant composition. Vaccines can be administered in the usual practice volume (in the range of 0.1 mL to 0.5 mL).

劑量定時取決於業內熟知因素。在初始投與後,可投與一或多次額外劑量以維持及/或強化抗體效價及T細胞免疫力。可能需要額外強化以維持免疫反應之保護程度,該等程度係以抗體效價及T細胞免疫力(例如ELISPOT)反映。此等免疫反應之程度係臨床研究標的。 Dose timing depends on factors well known in the art. After initial administration, one or more additional doses may be administered to maintain and/or enhance antibody titer and T cell immunity. Additional enhancement may be required to maintain the degree of protection of the immune response, as reflected by antibody titers and T cell immunity (eg, ELISPOT). The extent of these immune responses is the subject of clinical research.

對於組合疫苗接種而言,各免疫原可一起以一種組合物或單獨地以不同組合物投與。本文所述rdCMV係與一或多種期望免疫原同時投與。術語「同時」並不限於恰好在相同時間投與治療劑,而是,其意指,本文所述rdCMV及其他期望免疫原係以一定序列且在一定時間間隔內投與個體,以使得其可一起作用,從而提供與在其以其他方式投與時相比有所增加之益處。例如,各治療劑可在相同時間或以任一順序在不同時間點依序投與;然而,若不在相同時間投與,則其應在時間上充分靠近投與以提供期望治療效應。各治療劑可單獨地、以任一合適形式及藉由任何適宜途徑投與。 For combination vaccination, each immunogen can be administered together in one composition or separately in different compositions. The rdCMV lines described herein are administered concurrently with one or more desired immunogens. The term "simultaneously" is not limited to the administration of a therapeutic agent at exactly the same time, but rather means that the rdCMV and other desired immunogenic lines described herein are administered to the individual in a sequence and at intervals of time such that they are One works to provide an added benefit compared to when it is otherwise administered. For example, each therapeutic agent can be administered sequentially at the same time or in any order at different time points; however, if not administered at the same time, it should be sufficiently close in time to provide the desired therapeutic effect. Each therapeutic agent can be administered separately, in any suitable form, and by any suitable route.

患者群體Patient population

「患者」係指能經CMV感染之哺乳動物。在較佳實施例中,患者係人類。患者可經預防性或治療性治療。預防性 治療提供足夠保護性免疫力以降低CMV感染之可能性或嚴重程度,該感染包括原發性感染、復發性感染(即,彼等源於潛伏CMV再活化者)及再感染(即,彼等源於與患者先前所經歷者不同株CMV之感染者)。可執行治療性治療以降低CMV感染之嚴重程度或降低復發性或再感染之可能性/嚴重程度。 "Patient" means a mammal that can be infected with CMV. In a preferred embodiment, the patient is a human. Patients can be treated prophylactically or therapeutically. Preventive Treatment provides sufficient protective immunity to reduce the likelihood or severity of CMV infection, including primary infections, recurrent infections (ie, those from latent CMV reactivation) and reinfection (ie, they Originated from an infected person with a different strain of CMV than the patient previously experienced). Therapeutic treatment can be performed to reduce the severity of CMV infection or to reduce the likelihood/severity of relapse or reinfection.

可使用包含本文所述rdCMV之醫藥組合物執行治療。可將醫藥組合物投與一般群體,尤其彼等CMV感染(原發性、復發性或再)風險增加者或CMV感染將尤其成問題者(例如免疫受損個體、移植患者或孕婦)。在一個實施例中,給育齡女性、尤其早期青春期女性接種疫苗以降低在懷孕期間CMV感染(原發性、復發性或再)之可能性。 Treatment can be performed using a pharmaceutical composition comprising rdCMV as described herein. Pharmaceutical compositions can be administered to a general population, particularly those with an increased risk of CMV infection (primary, recurrent, or re-) or CMV infections (e.g., immunocompromised individuals, transplant patients, or pregnant women). In one embodiment, women of childbearing age, particularly early adolescent women, are vaccinated to reduce the likelihood of CMV infection (primary, relapsing or re-) during pregnancy.

彼等需要治療者包括彼等已具有感染者,以及彼等傾向於感染者或期望降低感染可能性者。治療可改善與CMV感染相關之疾病症狀及/或縮短CMV感染之時長及/或嚴重程度,該感染包括因潛伏CMV再活化所致之感染。 Those who need treatment include those who have already become infected, and those who prefer to be infected or who are expected to reduce the likelihood of infection. Treatment may ameliorate the symptoms of the disease associated with CMV infection and/or shorten the duration and/or severity of the CMV infection, including infections caused by latent CMV reactivation.

CMV感染(原發性、復發性或再)風險增加者包括免疫減弱患者或面對導致免疫力減弱之療法(例如,經受用於癌症之化學療法或輻射療法或服用免疫抑制藥物)之患者。本文所用「免疫力減弱」係指因不適當地起作用或不以正常健康成人之程度起作用之免疫反應而不太能抵抗感染的免疫系統。免疫力減弱患者之實例係嬰兒、幼兒、老人、孕婦患者或患有影響免疫系統功能之疾病(例如HIV感染或AIDS)之患者。 Patients at increased risk of CMV infection (primary, recurrent, or re-) include immunocompromised patients or patients facing treatments that cause reduced immunity (eg, undergoing chemotherapy or radiation therapy for cancer or taking immunosuppressive drugs). As used herein, "immune immunity" refers to an immune system that is less resistant to infection by an immune response that does not function properly or that does not function as a normal healthy adult. Examples of patients with reduced immunity are infants, young children, elderly people, pregnant women, or patients with diseases that affect the function of the immune system (such as HIV infection or AIDS).

實例 Instance

下文提供實例以進一步闡釋本發明之不同特徵。該等實例亦闡釋實踐本發明之有用方法。該等實例並不限制所主張本發明。 Examples are provided below to further illustrate various features of the invention. These examples also illustrate useful methods of practicing the invention. These examples do not limit the claimed invention.

實例1:五聚gH複合物之恢復Example 1: Recovery of pentameric gH complex

構築感染性CMV細菌人工染色體純系,以使得編碼病毒粒子表現由組裝至gH/gL支架上之UL128、UL130及UL131組成之五聚gH複合物。 Infectious CMV bacterial artificial chromosomes were constructed such that the coding virions exhibited a pentameric gH complex consisting of UL128, UL130 and UL131 assembled onto a gH/gL scaffold.

CMV株AD 169株最初係自7歲女孩之增殖腺分離(Elek及Stern,1974,Lancet,1:1)。將病毒在若干類型之人類纖維母細胞中傳代58次以衰減該病毒(Neff等人,1979,Proc Soc Exp Biol Med,160:32),其中最後5次傳代係在WI-38人類纖維母細胞中進行。使用AD 169病毒之此經傳代變體(在此研究中稱為Merck AD 169,MAD 169)作為親代病毒來構築感染性BAC純系。親代病毒AD 169與經傳代變體病毒MAD 169二者均不表現UL131或五聚gH複合物。 The CMV strain AD 169 strain was originally isolated from the proliferating gland of a 7 year old girl (Elek and Stern, 1974, Lancet, 1:1). The virus was passaged 58 times in several types of human fibroblasts to attenuate the virus (Neff et al., 1979, Proc Soc Exp Biol Med, 160:32), with the last 5 passages in WI-38 human fibers. Performed in mother cells. This passaged variant of AD 169 virus (referred to as Merck AD 169, MAD 169 in this study) was used as a parental virus to construct an infectious BAC line. Both the parental virus AD 169 and the passaged variant virus MAD 169 do not exhibit a UL131 or pentameric gH complex.

使用MAD 169作為親代病毒來構築感染性細菌人工染色體(BAC)純系。BAC載體係分子工具,其允許遺傳操作大腸桿菌中之大尺寸DNA片段,例如CMV基因組(約230 Kb)。在緊隨US28開放閱讀框之終止密碼子之後插入BAC元件以及GFP標記物基因(在病毒基因組中之US28與US29 ORF之間),其中在片段兩端處建立LoxP位點(圖1A)。簡言之,合成兩側為兩個loxP位點之含有GFP表現盒之DNA片段及CMV US28-US29序列並將其選殖至pBeloBAC11載 體中。利用限制酶Pme I將BAC載體線性化,並將其與自經純化病毒粒子提取之MAD 169 DNA一起共轉染至MRC-5細胞中。藉由綠色螢光表現鑑別之重組變體係經噬菌斑純化的。在一輪擴增後,自受感染細胞提取環形形式之病毒基因組,且將其電穿孔至大腸桿菌DH10細胞中。藉由PCR篩選細菌菌落中US28及US29區之存在。藉由EcoR I、EcoR V、Hind III、Spe I及Bam HI限制分析進一步檢測候選純系。在篩選後,一種純系bMAD-GFP顯示與親代MAD 169病毒相同之限制模式。 The infectious bacteria artificial chromosome (BAC) pure line was constructed using MAD 169 as a parental virus. The BAC vector is a molecular tool that allows genetic manipulation of large size DNA fragments in E. coli, such as the CMV genome (about 230 Kb). The BAC element and the GFP marker gene (between US28 and US29 ORF in the viral genome) were inserted immediately following the stop codon of the US28 open reading frame, wherein LoxP sites were established at both ends of the fragment (Fig. 1A). Briefly, the DNA fragment containing the GFP expression cassette and the CMV US28-US29 sequence, which are flanked by two loxP sites, were synthesized and cloned into pBeloBAC11 In the body. The BAC vector was linearized with restriction enzyme Pme I and co-transfected into MRC-5 cells with MAD 169 DNA extracted from purified virions. The recombinant system identified by green fluorescent expression was purified by plaque. After one round of amplification, the viral genome in circular form was extracted from infected cells and electroporated into E. coli DH10 cells. The presence of US28 and US29 regions in bacterial colonies was screened by PCR. Candidate lines were further tested by EcoR I, EcoR V, Hind III, Spe I and Bam HI restriction assays. After screening, one pure line of bMAD-GFP showed the same restriction pattern as the parental MAD 169 virus.

在大腸桿菌中以遺傳方式修復導致MAD 169中之上皮向性缺陷的UL131之第一外顯子中之移碼突變(圖1B)。具體而言,缺失UL131基因中之7腺嘌呤核苷酸伸展中一個nt A(圖1B)。1 nt之缺失足以挽救因UL131及(由此)五聚gH複合物現在表現所致之上皮及內皮細胞向性。藉由ELISA及西方墨點(western blot)來確認表現(數據未顯示)。藉由LoxP/Cre重組去除BAC區段來進一步修飾此純系。在ARPE-19細胞、即人類視網膜色素上皮細胞(ATCC登錄號CRL-2302)中轉染BAC DNA以回收感染性病毒(圖1C)。所得感染性病毒稱為BAC源上皮向性MAD 169病毒(beMAD),其與MAD 169不同之處僅在於以下兩個基因座:(1)UL131 ORF,其中缺失單一腺嘌呤核苷酸;及(2)34 bp LoxP位點,其插入US28與US29 ORF之間(參見表2)。 The frameshift mutation in the first exon of UL131 that causes epithelial deficiencies in MAD 169 was genetically repaired in E. coli (Fig. IB). Specifically, one of the 7 adenine nucleotides in the UL131 gene was deleted and one nt A was absent (Fig. 1B). A 1 nt deletion is sufficient to rescue epithelial and endothelial cell tropism due to UL131 and (and thus) pentameric gH complexes. Performance was confirmed by ELISA and western blot (data not shown). This pure line was further modified by LoxP/Cre recombination to remove the BAC segment. BAC DNA was transfected into ARPE-19 cells, human retinal pigment epithelial cells (ATCC Accession No. CRL-2302) to recover infectious virus (Fig. 1C). The resulting infectious virus is referred to as the BAC-derived epithelial MAD 169 virus (beMAD), which differs from MAD 169 only in the following two loci: (1) the UL131 ORF in which a single adenine nucleotide is deleted; 2) A 34 bp LoxP site inserted between US28 and US29 ORF (see Table 2).

BAC純系beMAD之基因組經完全定序。beMAD之總體基 因組結構與以ATCC AD 169變體(GenBank登錄號X17403)報導者相同,該變體包括兩個獨特區,即長獨立區(UL)及短獨特區(US)。各獨特區包括兩個重複序列,即長端重複(TRL)-長內部重複(IRL)、短端重複(TRS)-短內部重複(IRS)。經傳代變體MAD 169及beMAD源病毒之生長動力學在人類纖維母細胞細胞系MRC-5細胞(ATCC登錄號CCL-171)中不可區別(數據未顯示)。由於在纖維母細胞細胞上生長無需gH複合物,因此MAD 169與beMAD之間之gH複合物表現差異不相關。 The genome of the BAC pure beMAD is completely sequenced. The overall basis of beMAD Since the group structure is identical to that reported by the ATCC AD 169 variant (GenBank Accession No. X17403), the variant includes two distinct regions, the Long Independent Region (UL) and the Short Unique Region (US). Each unique region includes two repeating sequences, namely long-end repeat (TRL)-long internal repeat (IRL), short-end repeat (TRS)-short internal repeat (IRS). The growth kinetics of the passaged variants MAD 169 and beMAD-derived viruses were indistinguishable in the human fibroblast cell line MRC-5 cells (ATCC Accession No. CCL-171) (data not shown). Since no gH complex is required for growth on fibroblast cells, the difference in gH complex performance between MAD 169 and beMAD is not relevant.

實例2:習用不活化方法對gH複合物之效應Example 2: Effect of conventional inactivation method on gH complex

研究兩種習用病毒不活化方法(γ-輻照及β-丙內酯(BPL))對表現gH之CMV之效應。 The effects of two conventional virus inactivation methods (gamma-irradiation and beta-propiolactone (BPL)) on CMV expressing gH were investigated.

對凍乾病毒粒子執行γ-輻照。使用保守凍乾循環(-50℃冷凍及在-35℃下初次乾燥約30 hr、之後在25℃下二次乾燥6 hr)將濃度為0.15 mg/mL之存於HNS(25 mM組胺酸、150 mM NaCl、9% w/v蔗糖,pH 6.0)中之重組CMV疫苗調配物凍乾,獲得乾燥粉末。將疫苗於3 mL玻璃小瓶中凍 乾,其中各小瓶中填充0.5 ml。在凍乾結束時,在氮環境中塞住小瓶並移出試樣,加標記,波紋化(crimped)且在-70℃下儲存直至γ輻照。將小瓶在Co輻照器下輻照期望輻照劑量。 Gamma-irradiation was performed on the lyophilized virions. Concentration of 0.15 mg/mL in HNS (25 mM histidine) using a conservative lyophilization cycle (freezing at -50 °C and initial drying at -35 °C for about 30 hr followed by secondary drying at 25 °C for 6 hr) The recombinant CMV vaccine formulation in 150 mM NaCl, 9% w/v sucrose, pH 6.0) was lyophilized to obtain a dry powder. Freeze the vaccine in a 3 mL glass vial Dry, each of which is filled with 0.5 ml. At the end of lyophilization, the vials were stoppered in a nitrogen atmosphere and the samples removed, labeled, crimped and stored at -70 °C until gamma irradiation. The vial was irradiated with the desired irradiation dose under a Co irradiator.

對於BPL處理而言,將BPL母液添加至在ARPE-19細胞上生長之粗製病毒培養上清液中,以達到0.01%或0.1%(v/v)之最終濃度。在不同時間點利用硫代硫酸鈉終止反應。隨後藉由超速離心純化經BPL處理之表現gH之CMV。 For BPL treatment, BPL stock was added to the crude virus culture supernatant grown on ARPE-19 cells to achieve a final concentration of 0.01% or 0.1% (v/v). The reaction was terminated with sodium thiosulfate at various time points. The BPL-treated CMV-expressing CMV was then purified by ultracentrifugation.

藉由在ARPE-19細胞中進行噬菌斑分析來測定兩種方法之不活化動力學。簡言之,準備存於PBS中之病毒試樣連續稀釋物且使用0.1 mL來接種已播種ARPE-19細胞之6孔板之各孔。在37℃下將板培育1 hr,隨後每孔添加6 mL含有0.5%瓊脂糖之上覆培養基。在37℃下將板培育18天。為顯現噬菌斑,將約0.5 mL 5 mg/mL MTT溶液(噻唑藍溴化四唑鎓,Sigma M5655)添加至各孔中。在37℃將板培育2-4 hr並在lightbox下對噬菌斑計數(圖2A及2C)。 The inactivation kinetics of the two methods were determined by plaque assay in ARPE-19 cells. Briefly, serial dilutions of virus samples in PBS were prepared and 0.1 mL was used to inoculate wells of 6-well plates in which ARPE-19 cells were seeded. The plates were incubated for 1 hr at 37 ° C, followed by the addition of 6 mL of 0.5% agarose overlying medium per well. The plates were incubated for 18 days at 37 °C. To visualize plaques, approximately 0.5 mL of 5 mg/mL MTT solution (thiazole blue tetrazolium bromide, Sigma M5655) was added to each well. Plates were incubated for 2-4 hr at 37 °C and plaques were counted under lightbox (Figures 2A and 2C).

藉由測定在小鼠中誘導之中和抗體效價來分析表現gH之不活化CMV之免疫原性。簡言之,在第0週及第3週利用2.5 μg CMV/劑量對雌性Balb/c小鼠(n=10)實施免疫。在第4週採集血清並評估針對病毒上皮進入之中和活性。中和效價(NT50)定義為與陰性對照相比使病毒上皮進入減少50%之血清稀釋度之倒數。來自小鼠免疫原性研究之結果顯示,兩種習用不活化方法對由表現gH之CMV誘導之中和抗體效價具有負效應(圖2B及2D)。NT50效價之降低與 藉由γ-輻照或BPL處理之持續時間相關。延長處理使表現五聚gH複合物之CMV在小鼠中之免疫原性方面更像親代AD 169 CMV。當在兔及恆河猴中測試利用γ-輻照或BPL不活化之疫苗時,觀察到類似結果(數據未顯示)。該等觀察顯示,在所選不活化條件下,五聚gH複合物對兩種不活化方法皆敏感。 The immunogenicity of the inactive CMV expressing gH was analyzed by measuring the induction of neutralizing antibody titers in mice. Briefly, female Balb/c mice (n=10) were immunized with 2.5 μg CMV/dose at weeks 0 and 3. Serum was collected at week 4 and assessed for neutralizing activity against viral epithelium. Neutralization titer (NT50) is defined as the reciprocal of serum dilution that reduces viral epithelial entry by 50% compared to the negative control. The results from mouse immunogenicity studies showed that two conventional inactivation methods had a negative effect on the induction of neutralizing antibody titers by CMV expressing gH (Figures 2B and 2D). Reduction of NT50 titer and The duration is related by gamma-irradiation or BPL treatment. Prolonged treatment made the CMV expressing the pentameric gH complex more similar to the parental AD 169 CMV in immunogenicity in mice. Similar results were observed when γ-irradiated or BPL-inactivated vaccines were tested in rabbits and rhesus monkeys (data not shown). These observations show that the pentameric gH complex is sensitive to both inactivation methods under the selected inactivation conditions.

實例3:FKBP-必需蛋白融合物之構築及篩選Example 3: Construction and screening of FKBP-essential protein fusions

使用經衰減AD 169株骨架來構築CMV,該骨架重新獲得其上皮向性,同時為條件式複製缺陷型。使用實例1中所述方法來恢復上皮向性。 The CMV was constructed using the attenuated AD 169 strain skeleton, which regained its epithelial tropism and was conditionally replicated. The method described in Example 1 was used to restore epithelial tropism.

基於兩種標準來選擇欲融合至FKBP衍生物之病毒蛋白。首先,藉由蛋白組學分析在CMV病毒粒子中檢測不到所關注蛋白(Varnum等人,2004,J.Virol.78:10960),由此降低將FKBP融合蛋白納入病毒中之可能性。其次,所關注蛋白對於組織培養物中之病毒複製至關重要。 Viral proteins to be fused to FKBP derivatives are selected based on two criteria. First, the protein of interest was not detected in the CMV virions by proteomic analysis (Varnum et al., 2004, J. Virol. 78: 10960), thereby reducing the possibility of incorporating the FKBP fusion protein into the virus. Second, the protein of interest is critical for viral replication in tissue culture.

使用beMAD作為親代病毒,將FKBP衍生物(SEQ ID NO:12)個別地融合至12種必需病毒蛋白,包括IE1/2(SEQ ID NO:1)、pUL37x1、pUL44、pUL51(SEQ ID NO:3)、pUL52(SEQ ID NO:5)、pUL53、pUL56、pUL77、pUL79(SEQ ID NO:7)、pUL84(SEQ ID NO:9)、pUL87及pUL105。亦構築具有兩種融合至FKBP之不同必需蛋白之病毒,其將IE1/2及UL51中之每一者融合至FKBP衍生物(具有去穩定之IE1/2及UL51之rdCMV之基因組示於SEQ ID NO:14中)。在構築後,將所有重組BAC DNA轉染至 ARPE-19細胞中,且在含有Shld-1之培養基中培養。 The FKBP derivative (SEQ ID NO: 12) was individually fused to 12 essential viral proteins using beMAD as the parental virus, including IE1/2 (SEQ ID NO: 1), pUL37x1, pUL44, pUL51 (SEQ ID NO: 3), pUL52 (SEQ ID NO: 5), pUL53, pUL56, pUL77, pUL79 (SEQ ID NO: 7), pUL84 (SEQ ID NO: 9), pUL87 and pUL105. Also constructed is a virus having two different essential proteins fused to FKBP, which fused each of IE1/2 and UL51 to a FKBP derivative (the genome of rdCMV with destabilized IE1/2 and UL51 is shown in SEQ ID) NO: 14)). After construction, transfect all recombinant BAC DNA to ARPE-19 cells were cultured in medium containing Shld-1.

檢測病毒生長對Shld-1之依賴性。在噬菌斑分析中以2 μM Shld-1容易挽救IE1/2、UL51、UL52、UL84、UL79及UL87融合病毒(數據未顯示)。UL37x 1、UL77及UL53病毒亦產生噬菌斑,但噬菌斑較小,且其與親代beMAD相比生長顯著較慢。將Shld-1濃度增加至10 μM不會顯著促進病毒生長(數據未顯示)。UL56及UL105融合物未恢復,從而表明對該等蛋白加標籤會破壞該等蛋白之功能或相鄰基因之表現。 The dependence of virus growth on Shld-1 was examined. IE1/2, UL51, UL52, UL84, UL79, and UL87 fusion viruses were easily rescued with 2 μM Shld-1 in plaque assays (data not shown). The plaques were also produced by the UL37x 1, UL77 and UL53 viruses, but the plaques were small and they grew significantly slower than the parental beMAD. Increasing the Shld-1 concentration to 10 μM did not significantly promote virus growth (data not shown). The UL56 and UL105 fusions were not restored, indicating that tagging these proteins would disrupt the function of the proteins or the performance of adjacent genes.

在其他實驗中使用不同濃度之Shld-1來進一步評價在存在或不存在Shld-1下之病毒複製。藉由表現gH之CMV感染ARPE-19細胞,其中MOI為0.01 pfu/ml,該CMV亦含有融合至必需蛋白之FKBP衍生物。在感染1小時後,用新鮮培養基將細胞洗滌兩次以自接種物去除Shld-1。然後將接種物添加至在含有0.05 μM、0.1 μM、0.5 μM或2 μM Shield-1之培養基中培養之ARPE-19細胞中。感染後7天,採集上清液中之無細胞子代病毒且將其滴定於補充有2 mM Shield-1之ARPE-19細胞上。藉由50%組織培養感染劑量(TCID50)分析測定病毒效價。簡言之,此稀釋分析量化殺傷50%受感染宿主所需病毒之量。平鋪ARPE-19細胞並添加病毒之連續稀釋物。在培育後,人工觀察並記錄各病毒稀釋度之細胞死亡(即受感染細胞)之百分比。使用結果來以數學方式計算TCID50。 Different concentrations of Shld-1 were used in other experiments to further evaluate viral replication in the presence or absence of Shld-1. ARPE-19 cells were infected by CMV expressing gH with an MOI of 0.01 pfu/ml, which also contained FKBP derivatives fused to the essential protein. One hour after infection, the cells were washed twice with fresh medium to remove Shld-1 from the inoculum. The inoculum was then added to ARPE-19 cells cultured in medium containing 0.05 μM, 0.1 μM, 0.5 μM or 2 μM Shield-1. Seven days after infection, the cell-free progeny virus in the supernatant was collected and titrated onto ARPE-19 cells supplemented with 2 mM Shield-1. Viral titers were determined by 50% tissue culture infectious dose (TCID50) analysis. Briefly, this dilution analysis quantifies the amount of virus required to kill 50% of infected hosts. ARPE-19 cells were plated and serial dilutions of the virus were added. After incubation, the percentage of cell death (i.e., infected cells) for each virus dilution was manually observed and recorded. Use the results to mathematically calculate TCID50.

如圖3中所示,所有含有FKBP融合物之CMV之有效複製 取決於Shield-1濃度,但程度有所不同。較低濃度之Shield-1通常降低子代病毒產生之效價。在具有單一融合物之病毒中,僅UL51及UL52之複製絕對需要Shield-1。其他具有單一融合物之病毒(IE1/2、UL84、UL79及UL87)可在Shield-1不存在下產生可檢測子代病毒。當FKBP衍生物融合至UL51或UL52時,調控最嚴格。 As shown in Figure 3, all valid copies of CMV containing FKBP fusions Depending on the Shield-1 concentration, but to a different extent. Lower concentrations of Shield-1 generally reduce the potency produced by progeny viruses. Of the viruses with a single fusion, only copies of UL51 and UL52 require Shield-1. Other viruses with a single fusion (IE1/2, UL84, UL79, and UL87) can produce detectable progeny viruses in the absence of Shield-1. When FKBP derivatives are fused to UL51 or UL52, the regulation is most stringent.

在2 μM Shld-1存在或Shld-1不存在下比較具有IE1/2、UL51、IE1/2-UL51融合物之病毒之生長動力學與親代beMAD病毒。如圖4中所示,在Shld-1存在下,單一或雙融合物之生長動力學與親代beMAD相當。然而,在Shld-1不存在下,僅IE1/2可複製,但速率比親代beMAD低且慢。 The growth kinetics of the virus with the IE1/2, UL51, IE1/2-UL51 fusions were compared to the parental beMAD virus in the presence of 2 μM Shld-1 or in the absence of Shld-1. As shown in Figure 4, the growth kinetics of single or double fusions were comparable to the parental beMAD in the presence of Shld-1. However, in the absence of Shld-1, only IE1/2 can replicate, but at a slower and slower rate than the parental beMAD.

亦以不同細胞類型測試對雙融合病毒中之病毒複製之控制嚴格性(圖5)。該等細胞包括人類臍帶靜脈細胞(HUVEC)、MRC-5纖維母細胞、主動脈平滑肌細胞(AoMC)、骨骼肌細胞(SKMC)及CCF-STTG1星形細胞瘤細胞。藉由IE1/2-UL51融合病毒感染細胞(以5 pfu/細胞之MOI感染之CCF-STTG1除外),其中MOI為0.01 pfu/細胞,且隨後在存在或不存在Shield-1之培養基中培育。所有細胞類型在Shield-1存在下皆能支持溶解性病毒複製。在Shield-1不存在下未檢測到病毒產生。 The control rigor of viral replication in double fusion viruses was also tested in different cell types (Figure 5). Such cells include human umbilical vein cells (HUVEC), MRC-5 fibroblasts, aortic smooth muscle cells (AoMC), skeletal muscle cells (SKMC), and CCF-STTG1 astrocytoma cells. Cells were infected by IE1/2-UL51 fusion virus (except CCF-STTG1 infected with 5 pfu/cell MOI) with an MOI of 0.01 pfu/cell and subsequently incubated in medium with or without Shield-1. All cell types support soluble viral replication in the presence of Shield-1. No virus production was detected in the absence of Shield-1.

實例4:IE1/2-UL51雙融合病毒在動物中之免疫原性Example 4: Immunogenicity of IE1/2-UL51 double fusion virus in animals

在小鼠、兔及恆河猴中評估IE1/2-UL51雙融合病毒之免疫原性。首先在小鼠中比較針對IE1/2-UL51雙融合病毒或 親代beMAD病毒之劑量依賴性中和反應(圖6A)。在第0週及第4週利用劑量在0.12 μg至10 μg範圍內之beMAD或IE1/2-UL51雙融合病毒對6週齡雌性BALB/c小鼠實施免疫。第6週採集血清試樣並藉由CMV微中和分析於ARPE-19細胞上分析,如先前所述(Tang等人,Vaccine,「A novel throughput neutralization assay for supporting clinical evaluations of human cytomegalovirus vaccines」,2011年8月30日以電子形式發表,doi:10.1016/j.vaccine.2011.08.086)。在0.12 μg、0.37 μg、1.1 μg、3.3 μg及10 μg之劑量下比較反應。在低劑量範圍(0.12 μg至1.1 μg)下,beMAD之免疫原性略高,且在劑量量高於0.37 μg時始終檢測到中和抗體。在高劑量範圍(3.3 μg及10 μg)下,由兩種病毒誘導之中和抗體效價相當。 The immunogenicity of the IE1/2-UL51 double fusion virus was evaluated in mice, rabbits and rhesus monkeys. First compare the IE1/2-UL51 double fusion virus in mice or A dose-dependent neutralization reaction of the parental beMAD virus (Fig. 6A). 6-week-old female BALB/c mice were immunized at week 0 and week 4 with beMAD or IE1/2-UL51 double fusion virus at doses ranging from 0.12 μg to 10 μg. Serum samples were taken at week 6 and analyzed on ARPE-19 cells by CMV microneutralization assay as previously described (Tang et al., Vaccine, "A novel throughput neutralization assay for supporting clinical evaluations of human cytomegalovirus vaccines", Published on August 30, 2011 in electronic format, doi: 10.1016/j.vaccine.2011.08.086). The reactions were compared at doses of 0.12 μg, 0.37 μg, 1.1 μg, 3.3 μg, and 10 μg. At low doses (0.12 μg to 1.1 μg), beMAD is slightly more immunogenic and neutralizing antibodies are always detected at doses above 0.37 μg. In the high dose range (3.3 μg and 10 μg), the neutralization antibody titers induced by the two viruses were comparable.

接下來,比較劑量為10 μg之不同病毒在兔中之免疫原性。在第0週、第3週及第8週利用10 μg beMAD或所示融合病毒對雌性NZW兔實施免疫。採集第10週血清並藉由CMV微中和分析於ARPE-19細胞上分析(圖5B)。beMAD、單一融合病毒IE1/2或UL51及雙融合病毒IE1/2-UL51可顯著誘導高於MAD 169之中和抗體效價,該MAD 169係與AD 169類似且缺乏五聚gH複合物之病毒。此確認,藉由病毒表現gH複合物顯著增加重組CMV之免疫原性。 Next, the immunogenicity of different viruses at a dose of 10 μg in rabbits was compared. Female NZW rabbits were immunized with 10 μg of beMAD or the indicated fusion virus at weeks 0, 3 and 8. Serum at week 10 was collected and analyzed on ARPE-19 cells by CMV microneutralization assay (Fig. 5B). beMAD, single fusion virus IE1/2 or UL51 and double fusion virus IE1/2-UL51 significantly induced higher than MAD 169 neutralizing antibody titer, MAD 169 is a virus similar to AD 169 and lacking pentameric gH complex . This confirms that the expression of the gH complex by the virus significantly increases the immunogenicity of the recombinant CMV.

接下來,在恆河猴中測試100 μg雙融合IE1/2-UL51病毒或親代beMAD病毒之免疫原性。採集第12週血清並藉由CMV微中和分析於ARPE-19細胞上分析。第12週(第3次給 藥後)之GMT NT50效價分別係11500或15600。該等效價與自然感染個體中所觀察之NT50效價相當(圖5C)。 Next, the immunogenicity of 100 μg of the double-fused IE1/2-UL51 virus or the parental beMAD virus was tested in rhesus monkeys. Serum at week 12 was collected and analyzed on ARPE-19 cells by CMV microneutralization assay. Week 12 (3rd time The GMT NT50 titers of the drug are 11,500 or 15600, respectively. This equivalent price is comparable to the NT50 titer observed in naturally infected individuals (Fig. 5C).

在恆河猴中顯示雙融合病毒IE1/2-UL51 CMV疫苗誘導之免疫反應之壽命。動物接種疫苗10 μg/劑量或100 μg/劑量雙融合病毒IE1/2-UL51(基於總蛋白質量)。亦包括10 μg/劑量疫苗與非晶形鋁的羥基磷酸硫酸鹽(AAHS)或ISCOMATRIX®佐劑之調配物。於第0週、第8週及第24週在恆河猴(n=5)中投與疫苗。為比較,對照組於第0週、第4週及第24週接受30 μg/劑量之與MF59佐劑調配之重組gB。於縱軸上呈現所有組之NT50效價倒數的幾何平均值(GMT)(圖7)。在接種疫苗前,無任一猴中可檢測中和抗體效價>40。在第一次給藥後在第4週對於所有組檢測最低中和活性,中和抗體效價在約第12週及第28週(分別在第二次及第三次疫苗接種後4週)達到峰值。100 μg/劑量組在第28週之峰值GMT係14,500(約3倍高於10 μg/劑量組之4,660效價)。與10 μg/劑量組相比,ISCOMATRIX®佐劑而非AAHS提供佐劑益處。測得ISCOMATRIX®組在第28週之GMT為15,800,而AAHS組係3,000且10 μg/劑量組係4,660。檢測對照(gB/MF59)組之最低中和活性,其中峰值GMT決不超過200。在研究第72週,即在完成第0週、第8週及第24週之疫苗接種方案後接近1年,100 μg/劑量組及ISCOMATRIX®調配組之GMT分別維持在1400及3000。此時,10 μg/劑量組及AAHS組之GMT係約200。 The lifespan of the immune response induced by the double fusion virus IE1/2-UL51 CMV vaccine was shown in rhesus monkeys. Animals were vaccinated at 10 μg/dose or 100 μg/dose of double fusion virus IE1/2-UL51 (based on total protein amount). Formulations of 10 μg/dose vaccine with amorphous aluminum hydroxyphosphate sulfate (AAHS) or ISCOMATRIX® adjuvant are also included. Vaccines were administered in rhesus monkeys (n=5) at weeks 0, 8, and 24. For comparison, the control group received 30 μg/dose of recombinant gB formulated with MF59 adjuvant at week 0, week 4, and week 24. The geometric mean (GMT) of the reciprocal of NT50 titers for all groups is presented on the vertical axis (Figure 7). Neutralizing antibody titers >40 were not detected in any of the monkeys prior to vaccination. The lowest neutralizing activity was tested for all groups at week 4 after the first dose, neutralizing antibody titers at approximately week 12 and week 28 (4 weeks after the second and third vaccinations, respectively) Reach the peak. The peak GMT of the 100 μg/dose group at week 28 was 14,500 (approximately 3 times higher than the 4,660 titer of the 10 μg/dose group). The ISCOMATRIX® adjuvant, rather than AAHS, provides adjuvant benefit compared to the 10 μg/dose group. The ISCOMATRIX® group had a GMT of 15,800 at week 28, while the AAHS group was 3,000 and the 10 μg/dose group was 4,660. The lowest neutralizing activity of the control (gB/MF59) group was tested, with a peak GMT never exceeding 200. At the 72nd week of the study, nearly one year after the completion of the vaccination program at weeks 0, 8, and 24, the GMT of the 100 μg/dose group and the ISCOMATRIX® formulation group was maintained at 1400 and 3000, respectively. At this time, the GMT of the 10 μg/dose group and the AAHS group was about 200.

於疫苗接種方案之第28週(第3次給藥後4週)採集來自恆 河猴之周邊血單核細胞(PBMC)且在IFN-γ ELISPOT分析中評估。猴接種疫苗100 μg/劑量(圖8A)或10 μg/劑量(圖8B-8D)之雙融合病毒IE1/2-UL51。另外,不利用佐劑(圖8B)或利用AAHS(圖8C)或ISCOMATRIX®(圖8D)佐劑調配10 μg/劑量。使用代表五種HCMV抗原之經彙集上覆肽抗原來刺激IFN-γ之離體產生。所用HCMV抗原係IE1及IE2(兩種病毒調控蛋白)以及pp65、gB及pp 150(主要病毒結構抗原)。藉由ELISPOT反應之量級(幾何平均值)以及對病毒抗原之反應者比率來評價T細胞反應品質。在接種疫苗之前,任一猴中皆無抗原特異性ELISPOT效價(數據未顯示)。 Collected from the 28th week of the vaccination program (4 weeks after the third dose) from Heng Peripheral blood mononuclear cells (PBMC) were evaluated and evaluated in the IFN-γ ELISPOT assay. The monkey was vaccinated with a double fusion virus IE1/2-UL51 at 100 μg/dose (Fig. 8A) or 10 μg/dose (Fig. 8B-8D). In addition, 10 μg/dose was dispensed without adjuvant (Fig. 8B) or with AAHS (Fig. 8C) or ISCOMATRIX® (Fig. 8D) adjuvant. Ex vivo production of IFN-[gamma] was stimulated by pooling overlying peptide antigens representing five HCMV antigens. The HCMV antigens used were IE1 and IE2 (two viral regulatory proteins) and pp65, gB and pp 150 (major viral structural antigens). The quality of T cell responses was assessed by the magnitude of the ELISPOT reaction (geometric mean) and the ratio of responders to viral antigens. There was no antigen-specific ELISPOT titer in any of the monkeys prior to vaccination (data not shown).

在第28週,對五種HCMV抗原(即,IE1、IE2、pp65、gB及pp150)之ELISPOT反應之幾何平均值分別係186個、132個、253個、87個、257個斑點形成細胞(SFC)/106個PBMC(對於100 μg/劑量組而言)對21個、24個、107個、111個、33個SFC/106個PBMC(對於10 μg/劑量組而言)(圖8A及8B)。基於55個以上SFC/106個PBMC及抗原特異性反應相對於二甲基亞碸(DMSO)反應上升3倍以上之截止標準對各組(n=5)中之反應者進行評分。對五種HCMV抗原(即,IE1、IE2、pp65、gB及pp 150)之反應者數目係4個、4個、5個、1個、3個(對於100 μg/劑量組而言)對1個、1個、5個、4個、0個(對於10 μg/劑量組而言)。 At week 28, the geometric mean of the ELISPOT responses to the five HCMV antigens (ie, IE1, IE2, pp65, gB, and pp150) were 186, 132, 253, 87, and 257 spotted cells, respectively. SFC)/10 6 PBMCs (for 100 μg/dose group) versus 21, 24, 107, 111, 33 SFC/10 6 PBMCs (for 10 μg/dose group) 8A and 8B). Responders in each group (n=5) were scored based on a cut-off criterion of more than 55 SFC/10 6 PBMCs and an antigen-specific reaction that was more than 3-fold higher than the dimethyl hydrazine (DMSO) reaction. The number of responders to the five HCMV antigens (ie, IE1, IE2, pp65, gB, and pp 150) was 4, 4, 5, 1, and 3 (for the 100 μg/dose group) versus 1 1, 5, 4, 0 (for the 10 μg/dose group).

ISCOMATRIX®佐劑對針對10 μg/劑量雙融合病毒IE1/2-UL51之T細胞反應之效應顯示於圖8D中。對五種HCMV抗 原(即,IE1、IE2、pp65、gB及pp 150)之ELISPOT反應之幾何平均值分別係114個、53個、491個、85個、113個SFC/106個PBMC,且組(n=5)中之反應者數目分別係3個、2個、5個、3個、3個。具有ISCOMATRIX®佐劑之組中T細胞反應之量級及幅度與彼等100 μg/劑量組中者類似。 The effect of ISCOMATRIX® adjuvant on T cell responses to 10 μg/dose double fusion virus IE1/2-UL51 is shown in Figure 8D. The geometric mean values of the ELISPOT reactions for the five HCMV antigens (ie, IE1, IE2, pp65, gB, and pp 150) were 114, 53, 491, 85, 113 SFC/10 6 PBMCs, respectively. The number of responders in the group (n=5) was 3, 2, 5, 3, and 3, respectively. The magnitude and magnitude of T cell responses in the group with ISCOMATRIX® adjuvant were similar to those in the 100 μg/dose group.

在用HCMV抗原(pp65、IE1、IE2或完整HCMV病毒粒子)刺激後,在細胞內細胞介素染色中進一步分析用10 μg/劑量或100 μg/劑量具有ISCOMATRIX®之雙融合病毒IE1/2-UL51(基於總蛋白質量)接種之動物的PBMC。陰性對照係一種未接種疫苗雙融合病毒IE1/2-UL51之未經處理之猴,而陽性對照係葡萄球菌腸毒素B(SEB)。圖9顯示,陰性對照顯示對所有抗原刺激之最低反應,但對陽性對照劑葡萄球菌腸毒素B(SEB)有反應,如所預計。來自兩組之所有10只接種疫苗之猴皆以類似量級及模式對HCMV特異性抗原有反應。計算所有10隻猴對各抗原之幾何平均值。當用CMV抗原肽彙集物(即,pp65、IE1及IE2)刺激所有猴之PBMC時,其皆顯示相當CD8+(圖9A)及CD4+(圖9B)T細胞反應;但當用完整HCMV病毒粒子刺激時,其優先顯示CD4+ T細胞反應。此並不意外,此乃因完整病毒粒子係蛋白抗原且可能作為外源抗原處理且較佳藉由MHC II類分子呈遞至CD4+ T細胞。雙融合病毒IE1/2-UL51可誘發CD4+及CD8+表型二者之T細胞反應,此與彼等通常在具有HCMV感染之健康個體中所觀察者類似。 After stimulation with HCMV antigen (pp65, IE1, IE2 or whole HCMV virions), further analysis of intracellular interleukin staining with 10 μg/dose or 100 μg/dose with ISCOMATRIX® double fusion virus IE1/2- PBMC of animals vaccinated with UL51 (based on total protein amount). The negative control was an untreated monkey of the unvaccinated double fusion virus IE1/2-UL51, while the positive control was staphylococcal enterotoxin B (SEB). Figure 9 shows that the negative control showed the lowest response to all antigen stimulation but responded to the positive control staphylococcal enterotoxin B (SEB) as expected. All 10 vaccinated monkeys from both groups responded to HCMV-specific antigens in similar magnitudes and patterns. The geometric mean of each of the 10 monkeys for each antigen was calculated. When all of the monkey PBMCs were stimulated with the CMV antigen peptide pool (i.e., pp65, IE1, and IE2), they all showed comparable CD8+ (Fig. 9A) and CD4+ (Fig. 9B) T cell responses; but when stimulated with intact HCMV virions When it is preferentially displayed, the CD4+ T cell response is shown. This is not surprising, as it is due to intact virion protein antigens and may be treated as a foreign antigen and is preferably presented to CD4+ T cells by MHC class II molecules. The double fusion virus IE1/2-UL51 induces a T cell response in both the CD4+ and CD8+ phenotypes, similar to those typically observed in healthy individuals with HCMV infection.

比較雙融合病毒IE1/2-UL51與鋁鹽之不同調配物在恆河 猴中產生中和抗體之能力(圖10)。用HNS(鹼性緩衝液)、非晶形鋁的羥基磷酸硫酸鹽(AAHS)或Merck磷酸鋁佐劑(MAPA)調配30 μg/劑量雙融合病毒IE1/2-UL51且於第0週及第8週投與。於第12週採集之血清試樣顯示,儘管MAPA增強中和抗體誘導,但增強在統計學上不顯著(雙尾非成對t測試)。 Comparison of different formulations of double fusion virus IE1/2-UL51 and aluminum salt in Ganges The ability to produce neutralizing antibodies in monkeys (Figure 10). 30 μg/dose of double fusion virus IE1/2-UL51 was formulated with HNS (alkaline buffer), amorphous aluminum hydroxyphosphate sulfate (AAHS) or Merck aluminum phosphate adjuvant (MAPA) and at week 0 and 8 Weekly vote. Serum samples collected at week 12 showed that although MAPA enhanced neutralization antibody induction, the increase was not statistically significant (two-tailed unpaired t test).

實例5:儲存用緩衝液之鑑別Example 5: Identification of storage buffer

用合適緩衝液將存於HBSS(漢克氏平衡鹽溶液(Hank's Balanced Salt Solution))中且在-70℃下儲存直至使用之CMV病毒稀釋約10×。存於各試樣中之HBSS緩衝液之殘餘組份包括0.533 mM氯化鉀、0.044 mM磷酸二氫鉀、0.034 mM磷酸氫二鈉、13.79 mM氯化鈉、0.417 mM碳酸氫鈉及0.1% w/v葡萄糖。然後將試樣在室溫或2℃與8℃之間之溫度下儲存4天或冷凍解凍。對於冷凍-解凍而言,將試樣在-70℃下儲存至少1小時且在RT下解凍30分鐘,持續一或三次循環。在第4天使用病毒進入分析測試試樣之穩定性。簡言之,使用若干不同試樣稀釋執行分析以獲得反應曲線,且藉由非線性曲線擬合自病毒進入分析結果獲得EC50(μg/mL)值。較低EC50值代表較佳穩定性。比較穩定性試樣與-70℃冷凍對照試樣之EC50值。 Store in HBSS (Hank's Balanced Salt Solution) with a suitable buffer and store at -70 °C until the CMV virus used is diluted approximately 10×. The residual components of the HBSS buffer in each sample included 0.533 mM potassium chloride, 0.044 mM potassium dihydrogen phosphate, 0.034 mM disodium hydrogen phosphate, 13.79 mM sodium chloride, 0.417 mM sodium bicarbonate, and 0.1% w. /v glucose. The sample is then stored at room temperature or at a temperature between 2 ° C and 8 ° C for 4 days or frozen and thawed. For freeze-thaw, the samples were stored at -70 ° C for at least 1 hour and thawed at RT for 30 minutes for one or three cycles. The stability of the test sample was tested using virus entry on day 4. Briefly, analysis was performed using several different sample dilutions to obtain a reaction curve, and EC50 (μg/mL) values were obtained from the virus entry analysis results by non-linear curve fitting. Lower EC50 values represent better stability. The EC50 values of the stability samples and the -70 °C frozen control samples were compared.

病毒進入分析量測CMV感染ARPE-19細胞及表現IE1(即早期蛋白1)之能力。在透明96孔板中執行分析。使用IE1特異性一級抗體及生物素化二級抗體來檢測固定細胞中之靶蛋白,且使用IR Dye 800CW抗生蛋白鏈菌素(Streptavidin) 以及Sapphire 700/DRAQ5(對於細胞輸入正規化)來量化各孔之螢光信號。將結果繪製為800/700積分強度比(Integrated Intensity Ratio,Integ.Ratio)對CMV濃度(總蛋白,μg/mL)。亦使用非線性曲線擬合自感染力分析結果獲得EC50值。由於ARPE-19細胞之病毒感染依賴病毒糖蛋白抗原、特定而言五聚gH複合物之完整性,因此EC50值反映病毒顆粒在該等條件下之保持程度。 Viral entry assays measure the ability of CMV to infect ARPE-19 cells and express IE1 (ie, early protein 1). Analysis was performed in a transparent 96-well plate. Detection of target proteins in fixed cells using IE1-specific primary antibodies and biotinylated secondary antibodies, and using IR Dye 800CW Streptavidin And Sapphire 700/DRAQ5 (for cell input normalization) to quantify the fluorescent signal of each well. The results were plotted as 800/700 Integrated Intensity Ratio (Integ. Ratio) versus CMV concentration (total protein, μg/mL). EC50 values were also obtained from the results of the infectivity analysis using a non-linear curve fit. Since the viral infection of ARPE-19 cells is dependent on the integrity of the viral glycoprotein antigen, in particular the pentameric gH complex, the EC50 value reflects the extent to which the viral particles are maintained under these conditions.

如圖11中所示,CMV當在RT下在HBSS中儲存4天時,損失感染力。此外,當藉由病毒進入分析評價時,在HBSS中進行3次冷凍-解凍循環導致感染力完全損失。因此,HBSS並非CMV儲存用最佳緩衝液。 As shown in Figure 11, the CMV lost its infectivity when stored in HBSS for 4 days at RT. Furthermore, when evaluated by virus entry analysis, three freeze-thaw cycles in HBSS resulted in complete loss of infectivity. Therefore, HBSS is not the optimal buffer for CMV storage.

使用3至8之pH範圍檢測pH對CMV室溫穩定性之效應。利用以下緩衝液:檸檬酸鹽緩衝液(25 mM)(pH 3.0);乙酸鹽緩衝液(25 mM)(pH 4);乙酸鹽緩衝液(25 mM)(pH 5);組胺酸緩衝液(25 mM)(pH 6);HEPES緩衝液(25 mM)(pH 7);漢克氏平衡鹽溶液(HBSS)(pH 7.5)及Tris緩衝液(25 mM)(pH 8)。 The effect of pH on CMV room temperature stability was examined using a pH range of 3 to 8. The following buffers were used: citrate buffer (25 mM) (pH 3.0); acetate buffer (25 mM) (pH 4); acetate buffer (25 mM) (pH 5); histidine buffer (25 mM) (pH 6); HEPES buffer (25 mM) (pH 7); Hank's Balanced Salt Solution (HBSS) (pH 7.5) and Tris buffer (25 mM) (pH 8).

藉由用合適緩衝液將病毒本體稀釋10次來準備試樣。將試樣在RT(25℃)下儲存4天。在第4天,藉由利用病毒進入分析來量測試樣之穩定性。將於-70℃下冷凍儲存之存於HBSS中之CMV作為對照處理。在0時及第4天獲得各試樣之UV-Vis譜以檢測在儲存期間發生之結構變化及聚集。 The sample was prepared by diluting the virus body 10 times with a suitable buffer. The samples were stored at RT (25 ° C) for 4 days. On day 4, the stability of the test sample was quantified by using virus entry analysis. The CMV stored in HBSS, which was stored frozen at -70 ° C, was treated as a control. The UV-Vis spectra of each sample were obtained at 0 and 4 days to detect structural changes and aggregation that occurred during storage.

如圖12中所示,與其他測試pH相比,25 mM組胺酸緩衝液(pH 6)藉由保留在RT下之較高感染力提供CMV之較佳穩 定性。UV譜之二階導數指示病毒在所有pH下之類似結構特徵(數據未顯示)。在任一測試pH下皆未觀察到顯著聚集,如藉由在350 nm下之光密度所量測(數據未顯示)。 As shown in Figure 12, 25 mM histidine buffer (pH 6) provides better stability of CMV by retaining higher infectivity at RT compared to other tested pH. Qualitative. The second derivative of the UV spectrum indicates similar structural features of the virus at all pH (data not shown). No significant aggregation was observed at any of the tested pHs, as measured by optical density at 350 nm (data not shown).

在25 mM組胺酸緩衝液(pH 6)中測試尿素單獨或與氯化鈉組合對CMV病毒穩定性之效應。單獨添加2%尿素對CMV穩定性不具有效應。然而,2%尿素與150 mM NaCl組合改良CMV在RT下之穩定性(圖13)。 The effect of urea alone or in combination with sodium chloride on the stability of CMV virus was tested in 25 mM histidine buffer (pH 6). The addition of 2% urea alone had no effect on CMV stability. However, the combination of 2% urea and 150 mM NaCl improved the stability of CMV at RT (Figure 13).

在pH 6下檢測離子強度對CMV穩定性之效應。將增加濃度之NaCl(0 mM、75 mM、150 mM及320 mM NaCl)添加至25 mM組胺酸緩衝液(pH 6)中。CMV穩定性取決於離子強度,其中較佳離子強度產生較佳穩定性(圖14)。尿素之存在對CMV穩定性不具有或具有最低效應(數據未顯示)。 The effect of ionic strength on CMV stability was examined at pH 6. Increasing concentrations of NaCl (0 mM, 75 mM, 150 mM, and 320 mM NaCl) were added to 25 mM histidine buffer (pH 6). CMV stability is dependent on ionic strength, with better ionic strength resulting in better stability (Figure 14). The presence of urea did not have or had the lowest effect on CMV stability (data not shown).

另外,篩選若干其他賦形劑(蔗糖、山梨醇、甘油及脯胺酸)對在室溫下表現gH之CMV穩定性之效應。在室溫下將所欲測試賦形劑添加至存於25 mm組胺酸緩衝液(pH 6)中之CMV中,且保持4天,之後使用病毒進入分析量測CMV病毒穩定性。計算試樣之EC50值。在所有所測試賦形劑中,150 mM NaCl單獨或與9% w/v蔗糖組合在pH 6下提供較佳穩定性(數據未顯示)。因此,所推薦CMV在RT下儲存用緩衝液係具有150 mM NaCl且具有或不具有9% w/v蔗糖之25 mM組胺酸(pH 6)。 In addition, several other excipients (sucrose, sorbitol, glycerol, and valine) were screened for the effect of CMV stability at g at room temperature. The test vehicle to be tested was added to CMV in 25 mm histidine buffer (pH 6) at room temperature for 4 days, after which CMV virus stability was measured using virus entry assay. Calculate the EC 50 value of the sample. Of all the tested excipients, 150 mM NaCl alone or in combination with 9% w/v sucrose provided better stability at pH 6 (data not shown). Therefore, the recommended CMV storage buffer has 25 mM histidine (pH 6) with 150 mM NaCl and with or without 9% w/v sucrose at RT.

研究在冷凍-解凍期間低溫保護劑對CMV穩定性之效應。如前文所述(圖11),當經受3次冷凍-解凍循環時,存於HBSS中之CMV完全損失其感染力。篩選若干低溫保護 劑(包括蔗糖、山梨醇、甘油)降低對CMV之冷凍-解凍脅迫之能力。對於各冷凍-解凍循環而言,將試樣在-70℃下冷凍至少1小時且在RT下解凍30分鐘。添加低溫保護劑可增加病毒之穩定性。此外,當與其他所測試低溫保護劑比較時,9% w/v蔗糖與150 mM氯化鈉組合可顯著增強病毒之穩定性(圖15)。因此,所推薦CMV在-70℃下儲存或至多3次冷凍-解凍循環用緩衝液組合物係25 mM組胺酸、150 mM NaCl及9%蔗糖(HNS緩衝液)。 The effect of cryoprotectants on CMV stability during freeze-thaw was investigated. As described above (Fig. 11), CMV stored in HBSS completely lost its infectivity when subjected to 3 freeze-thaw cycles. Screening for some low temperature protection Agents (including sucrose, sorbitol, glycerol) reduce the ability to freeze-thaw stress on CMV. For each freeze-thaw cycle, the samples were frozen at -70 °C for at least 1 hour and thawed at RT for 30 minutes. Adding a cryoprotectant increases the stability of the virus. In addition, the combination of 9% w/v sucrose and 150 mM sodium chloride significantly enhanced the stability of the virus when compared to other tested cryoprotectants (Figure 15). Therefore, the recommended CMV is stored at -70 ° C or up to 3 freeze-thaw cycles with a buffer composition of 25 mM histidine, 150 mM NaCl and 9% sucrose (HNS buffer).

針對在3次冷凍-解凍循環、冷藏(2-8℃)及RT(25℃)期間對CMV穩定性之保護,比較HNS緩衝液與HBSS緩衝液。HNS緩衝液提供CMV活病毒在所有測試儲存條件下之較佳穩定性(數據未顯示)。 HNS buffer and HBSS buffer were compared for protection against CMV stability during 3 freeze-thaw cycles, refrigeration (2-8 °C) and RT (25 °C). HNS buffer provided better stability of CMV live virus under all test storage conditions (data not shown).

實例6:CMV在HNS緩衝液中之穩定性Example 6: Stability of CMV in HNS Buffer

在HNS緩衝液中供應雙融合IE1/2-UL51 CMV病毒母液且將其儲存在-70℃下直至使用。在100 μg/mL之濃度(基於藉由Bradford分析量測之總蛋白含量)下執行穩定性研究。用HNS緩衝液稀釋本體病毒以獲得最終病毒濃度。然後將試樣儲存在合適溫度下且如針對所述進行測試至多3個月。對於冷凍-解凍而言,將試樣在-70℃下冷凍至少1小時且在室溫下解凍30分鐘。在不同時間點取出(pulled)試樣且將其保持在-70℃下冷凍儲存直至分析。 The double-fused IE1/2-UL51 CMV virus stock was supplied in HNS buffer and stored at -70 °C until use. Stability studies were performed at a concentration of 100 μg/mL based on total protein content as measured by Bradford analysis. The bulk virus was diluted with HNS buffer to obtain the final virus concentration. The samples were then stored at the appropriate temperature and tested as described for up to 3 months. For freeze-thaw, the samples were frozen at -70 °C for at least 1 hour and thawed at room temperature for 30 minutes. Samples were pulled at different time points and kept frozen at -70 °C until analysis.

使用Bradford分析來量測試樣之總蛋白含量。試樣之總蛋白含量在3個月時段內無變化(數據未顯示)。 The total protein content of the test samples was quantified using Bradford analysis. The total protein content of the samples did not change over the 3 month period (data not shown).

藉由使用DLS方法量測試樣之流體動力學直徑來監測試 樣中之CMV隨時間之粒徑。此方法監測病毒顆粒隨時間及在不同儲存溫度下之任何聚集或破壞。未觀察到真正趨勢,其中某些試樣之粒徑有偶發變化(數據未顯示)。結果指示,病毒顆粒完整且在升高溫度下不聚集。 Monitor the test by measuring the hydrodynamic diameter of the sample using the DLS method The particle size of the CMV in the sample over time. This method monitors any accumulation or disruption of viral particles over time and at different storage temperatures. No real trend was observed, with some samples having sporadic changes in particle size (data not shown). The results indicate that the virions are intact and do not aggregate at elevated temperatures.

實例7:儲存條件對病毒進入及免疫原性之效應Example 7: Effect of storage conditions on viral entry and immunogenicity

藉由使CMV試樣經受不同儲存溫度來觀察病毒進入效價(EC50值)之顯著變化(數據未顯示)。與2-8℃及25℃相比,在-20℃下儲存導致較低病毒進入效價。發現2-8℃試樣之效價之病毒進入效價低於25℃儲存。基於EC50值,按以下順序對儲存溫度分級(自最穩定至最不穩定):25℃>2-8℃>-20℃,至多1個月時間點。對於在-20℃、2-8℃及25℃下儲存之試樣而言,在3個月時間點處不可檢測病毒進入效價。 Significant changes in virus entry titers (EC50 values) were observed by subjecting the CMV samples to different storage temperatures (data not shown). Storage at -20 °C resulted in lower virus entry titers compared to 2-8 °C and 25 °C. It was found that the virus at a potency of 2-8 ° C sample entered the titer below 25 ° C for storage. Based on the EC50 values, the storage temperature was graded (from most stable to most unstable) in the following order: 25 °C > 2-8 °C > -20 °C, up to 1 month time point. For samples stored at -20 ° C, 2-8 ° C, and 25 ° C, the virus entry titer was not detectable at the 3 month time point.

在穩定性研究結束時起始小鼠免疫原性研究以測定儲存溫度對CMV誘導CMV中和抗體之能力之效應。在第0天用2.5 μg/劑量疫苗經肌內對小鼠實施免疫且在第21天實施強化,之後在第28天抽血。使用ARPE 19細胞測試小鼠血清中針對表現gH之CMV之中和抗體且藉由非線性曲線擬合獲得NT50效價。 Mouse immunogenicity studies were initiated at the end of the stability study to determine the effect of storage temperature on the ability of CMV to induce CMV neutralizing antibodies. Mice were immunized intramuscularly with a 2.5 μg/dose vaccine on day 0 and intensified on day 21, after which blood was drawn on day 28. The CMV neutralizing antibodies against gH in mouse serum were tested using ARPE 19 cells and NT50 titers were obtained by non-linear curve fitting.

評估在不同溫度下儲存3個月對IE1/2-UL51雙融合CMV免疫原性之效應。NT50效價取決於儲存溫度,其中與-70℃冷凍對照相比,更高溫度降低效價,但不顯著(p=0.2584,單因子ANOVA)(圖16A)。在-20℃下儲存之調配物之NT50效價較低,小於1/2,但與-70℃冷凍對照相比,該等試樣之病毒進入分析效價受到顯著影響。-20℃、2-8℃及25℃ 穩定性試樣之NT50效價之趨勢遵循針對該等試樣獲得之CMV質量ELISA效價。 The effect of storage at different temperatures for 3 months on the immunogenicity of IE1/2-UL51 double fusion CMV was assessed. The NT50 titer was dependent on the storage temperature, with higher temperatures lowering the potency compared to the -70 °C frozen control, but not significant (p = 0.2584, one-way ANOVA) (Figure 16A). The NT50 titers of the formulations stored at -20 °C were lower, less than 1/2, but the virus entry analysis titers of these samples were significantly affected compared to the -70 °C frozen control. -20 ° C, 2-8 ° C and 25 ° C The trend of NT50 titers for stability samples follows the CMV quality ELISA titers obtained for these samples.

評估在解凍後在不同溫度下儲存8小時對IE1/2-UL51雙融合CMV免疫原性之效應。比較該等調配物與-70℃冷凍對照之NT50效價。將試樣在任一測試溫度下儲存8小時不影響NT50效價(p=0.5865,單因子ANOVA)(圖16B)。 The effect of storage at different temperatures for 8 hours after thawing on the immunogenicity of IE1/2-UL51 double fusion CMV was assessed. The NT50 titers of the formulations were compared to the -70 °C frozen control. Storage of the samples at any of the test temperatures for 8 hours did not affect NT50 titers (p = 0.5865, one-way ANOVA) (Figure 16B).

在小鼠免疫原性研究中評估在將試樣解凍後雙融合IE1/2-UL51 CMV在25℃下儲存不同時間點之效應。比較該等調配物與-70℃冷凍對照之NT50效價。將試樣在25℃下存儲長達一週不影響NT50效價(p=0.1848,未成對雙尾t測試)。3個月時,NT 50效價下降到略低於1/2,從而指示調配物在25℃下保持較長時間時可能有穩定性問題(數據未顯示)。 The effect of double-fusion of IE1/2-UL51 CMV at 25 ° C for different time points after thawing the samples was evaluated in a mouse immunogenicity study. The NT50 titers of the formulations were compared to the -70 °C frozen control. Storage of the sample at 25 ° C for up to one week did not affect the NT50 titer (p = 0.1848, unpaired two-tailed t test). At 3 months, the NT 50 titer decreased to slightly below 1/2, indicating that the formulation may have stability problems at 25 ° C for a longer period of time (data not shown).

藉由小鼠免疫原性來評估3次冷凍-解凍循環對在HNS緩衝液中調配之雙融合IE1/2-UL51 CMV的效應。與-70℃冷凍對照相比,雙融合CMV調配物之3次冷凍-解凍(F/T)循環不會影響免疫原性(p=0.2103,未成對雙尾t測試)(數據未顯示)。 The effect of three freeze-thaw cycles on the double-fused IE1/2-UL51 CMV formulated in HNS buffer was evaluated by mouse immunogenicity. The 3 freeze-thaw (F/T) cycles of the double-fusion CMV formulation did not affect immunogenicity compared to the -70 °C frozen control (p=0.2103, unpaired two-tailed t test) (data not shown).

其他實施例在下文申請專利範圍內。儘管已顯示並闡述若干實施例,但可作出各種修改,此並不背離本發明之精神及範圍。 Other embodiments are within the scope of the following patent application. While a number of embodiments have been shown and described, various modifications may be made without departing from the spirit and scope of the invention.

圖1A-1B顯示恢復五聚gH複合物表現之CMV株之構築示意圖。(A)產生可自切除細菌人工染色體(BAC)以操作AD 169病毒基因組之策略。(B)修復UL131中之移碼突變以恢復其表現。(C)用cre重組酶基因替代GFP以建立可自切除CMV BAC。 1A-1B show schematic diagrams showing the construction of a CMV strain that restores the performance of the pentameric gH complex. (A) Producing a self-removing bacterial artificial chromosome (BAC) to manipulate AD 169 viral genome strategy. (B) Repair the frameshift mutation in UL131 to restore its performance. (C) Replacing GFP with the cre recombinase gene to establish a self-cleavable CMV BAC.

圖2A-2D顯示習用不活化方法對gH複合物免疫原性之效應。使用γ-輻照(A、B)及β-丙內酯E(BPL)(C、D)來使表現gH複合物之CMV不活化。不活化動力學係藉由噬菌斑分析測定(A、C),而免疫原性係藉由評估來自投與CMV之小鼠之血清針對病毒上皮細胞進入之中和活性來測定(B、D)。 Figures 2A-2D show the effect of the conventional inactivation method on the immunogenicity of the gH complex. Γ-irradiation (A, B) and β-propiolactone E (BPL) (C, D) were used to deactivate CMV expressing the gH complex. The inactivation kinetics were determined by plaque assay (A, C), and the immunogenicity was determined by assessing the entry of neutralizing activity against viral epithelial cells by serum from mice administered with CMV (B, D). ).

圖3顯示具有各種融合至FKBP衍生物之必需蛋白之表現gH複合物之CMV的Shield 1濃度依賴性子代病毒產生。ARPE-19細胞以0.01 PFU/細胞之複數(multiplicity)經rdCMV病毒感染1 h,用新鮮培養基洗滌兩次,且在含有0 μM、0.05 μM、0.1 μM、0.5 μM或2 μM Shield-1之生長培養基中培育。感染後7天,採集無細胞病毒,且藉由在2 μM Shield 1存在下對ARPE-19細胞實施TCID50分析來測定病毒效價。 Figure 3 shows Shield 1 concentration-dependent progeny virus production of CMVs of the gH complexes with various essential proteins fused to FKBP derivatives. ARPE-19 cells were infected with rdCMV virus for 1 h at 0.01 PFU/cell multiplicity, washed twice with fresh medium, and grown with 0 μM, 0.05 μM, 0.1 μM, 0.5 μM or 2 μM Shield-1 Cultivate in the medium. Seven days after infection, cell-free virus was collected and viral titer was determined by performing TCID50 analysis on ARPE-19 cells in the presence of 2 μM Shield 1.

圖4A-4D顯示rdCMV在ARPE-19細胞中之生長動力學。細胞以0.01 PFU/細胞之複數經含有(A)IE1/2、(B)UL51、(C)IE1/2-UL51融合蛋白之病毒或(D)親代beMAD病毒感染。在1小時後,用新鮮培養基將細胞洗滌兩次,且在Shield-1不存在(空心圓)或2 μM Shield-1存在(實心圓)下培育。在感染後於所示時間點採集無細胞病毒,且藉由對含有2 μM Shield-1之培養基中之ARPE-19細胞實施TCID50分 析來量化感染力病毒。 Figures 4A-4D show the growth kinetics of rdCMV in ARPE-19 cells. The cells were infected with a virus containing (A) IE1/2, (B) UL51, (C) IE1/2-UL51 fusion protein or (D) parental beMAD virus at a dose of 0.01 PFU/cell. After 1 hour, the cells were washed twice with fresh medium and incubated in the absence of Shield-1 (open circles) or 2 μM Shield-1 (filled circles). Cell-free virus was collected at the indicated time points after infection, and TCID50 was performed on ARPE-19 cells in medium containing 2 μM Shield-1. Analyze to quantify infectious viruses.

圖5A-5E係IE1/2-UL51 rdCMV在不同細胞類型中之生長動力學。(A)MRC-5、(B)HUVEC、(C)AoSMC、(D)SKMC、(E)CCF-STTG1細胞經rdCMV病毒感染且培育1小時。用新鮮培養基將細胞洗滌兩次,且隨後在Shield-1不存在(空心圓)或2 μM Shield-1存在(實心圓)下培育。在感染後於所示時間點採集無細胞病毒,且藉由對含有2 μM Shield-1之培養基中之ARPE-19細胞實施TCID50分析來量化感染力病毒。 Figures 5A-5E are growth kinetics of IE1/2-UL51 rdCMV in different cell types. (A) MRC-5, (B) HUVEC, (C) AoSMC, (D) SKMC, (E) CCF-STTG1 cells were infected with rdCMV virus and incubated for 1 hour. The cells were washed twice with fresh medium and subsequently incubated in the absence of Shield-1 (open circles) or 2 μM Shield-1 (filled circles). Acellular virus was collected at the indicated time points after infection, and the infectious virus was quantified by performing TCID50 analysis on ARPE-19 cells in medium containing 2 μM Shield-1.

圖6A-6C係IE1/2-UL51 rdCMV在小鼠、兔及恆河猴中之免疫原性分析。(A)在第0週及第4週用beMAD(空心圓)或IE1/2-UL51 rdCMV(實心圓)對小鼠實施免疫。(B)在第0週、第3週及第8週用10 μg beMAD或所示rdCMV對兔實施免疫。(C)在第0週及第8週用100 μg beMAD或IE1/2-UL51rdCMV對恆河猴實施免疫。在各情形下,採集血清試樣且藉由對ARPE-19細胞實施CMV微中和分析來分析。線條指示在各組中之幾何平均中和效價(NT50)。 Figures 6A-6C are immunogenicity analyses of IE1/2-UL51 rdCMV in mice, rabbits, and rhesus monkeys. (A) Mice were immunized with beMAD (open circles) or IE1/2-UL51 rdCMV (closed circles) at weeks 0 and 4. (B) Rabbits were immunized with 10 μg of beMAD or the indicated rdCMV at week 0, week 3 and week 8. (C) Immunization of rhesus monkeys with 100 μg beMAD or IE1/2-UL51rdCMV at week 0 and week 8. In each case, serum samples were collected and analyzed by performing CMV microneutralization analysis on ARPE-19 cells. The lines indicate the geometric mean neutralization titer (NT50) in each group.

圖7顯示接種疫苗雙融合病毒IE1/2-UL51之恆河猴之縱軸中和效價。在第0週、第8週及第24週接種所示疫苗劑量或調配物之恆河猴組(n=5)(顯示為紅色三角形),而一個組在第0週、第4週及第24週接受gb/mf59(30 mg/劑量)。在所示時間點採集免疫血清並在病毒中和分析中進行評估。以該組之標準誤差在縱軸上繪製NT50效價之GMT。AAHS:非晶形鋁的羥基磷酸硫酸鹽;IMX: ISCOMATRIX;HNS:鹼性緩衝液。 Figure 7 shows the vertical axis neutralization titer of rhesus monkeys vaccinated with the double fusion virus IE1/2-UL51. At the 0th, 8th, and 24th week, the indicated doses or formulations of the rhesus monkey group (n=5) (shown as red triangles) were inoculated, while one group was at week 0, week 4, and Gb/mf59 (30 mg/dose) was received at 24 weeks. Immune sera were collected at the indicated time points and evaluated in virus neutralization assays. The NT50 of the NT50 titer was plotted on the vertical axis with the standard error of the set. AAHS: Amorphous aluminum hydroxyphosphate sulfate; IMX: ISCOMATRIX; HNS: alkaline buffer.

圖8A-8D顯示接種疫苗100 μg(A)或10 μg(B-D)/劑量雙融合病毒IE1/2-UL51之恆河猴中之IFN-γ ELISPOT。不使用佐劑(A-B),或使用AAHS(C)或ISCOMATRIX(D)。用代表HCMV抗原之肽彙集物刺激PBMC。灰條代表該組(n=5)之各抗原之GMT。各抗原之反應者比率於各圖內顯示於各抗原之頂部。 Figures 8A-8D show IFN-γ ELISPOT in rhesus monkeys vaccinated with 100 μg (A) or 10 μg (B-D) per dose of double fusion virus IE1/2-UL51. Do not use adjuvant (A-B), or use AAHS (C) or ISCOMATRIX (D). PBMCs were stimulated with a pool of peptides representing HCMV antigens. Gray bars represent the GMT of each antigen of this group (n=5). The responder ratio of each antigen is shown on the top of each antigen in each figure.

圖9A-9B顯示在恆河猴中接種疫苗雙融合病毒IE1/2-UL51能誘導CD8+(A)及CD4+(B)表型二者之T細胞反應。自給予100 μg或10 μg劑量具有作為佐劑之ISCOMATRIX®之疫苗之猴採集PBMC。用代表HCMV抗原之肽彙集物刺激PBMC,之後針對IFN-γ及CD4+/CD8+表面T細胞標記物進行染色。數據呈現為每百萬PBMC中CD4+/CD8+陽性、IFN-γ陽性細胞之數目。線條代表接受相同疫苗之組(n=5)之幾何平均值(GMT)。各圖底部之數字代表兩個接種疫苗組(n=10)之GMT。CMV:經純化病毒;SEB:用作陽性對照劑之細胞分裂促進劑;IMX:ISCOMATRIX。 Figures 9A-9B show that vaccination of the double fusion virus IE1/2-UL51 in rhesus monkeys induces a T cell response in both the CD8+ (A) and CD4+ (B) phenotypes. PBMC were harvested from monkeys given a vaccine of ISCOMATRIX® as an adjuvant at a dose of 100 μg or 10 μg. PBMCs were stimulated with a pool of peptides representing HCMV antigens, followed by staining for IFN-[gamma] and CD4+/CD8+ surface T cell markers. Data are presented as the number of CD4+/CD8+ positive, IFN-[gamma] positive cells per million PBMC. Lines represent the geometric mean (GMT) of the group receiving the same vaccine (n=5). The numbers at the bottom of each figure represent the GMT of two vaccinated groups (n=10). CMV: purified virus; SEB: cell division promoter used as a positive control; IMX: ISCOMATRIX.

圖10顯示,Merck磷酸鋁佐劑(MAPA)可提高在猴中之中和抗體效價。在第0週及第8週用30 μg劑量在HNS(鹼性緩衝液)、AAHS或MAPA中調配之雙融合病毒疫苗對恆河猴實施免疫。在第12週採集血清試樣且評估中和效價。線條代表該組之幾何平均值。 Figure 10 shows that Merck Aluminium Phosphate Adjuvant (MAPA) increases neutralizing antibody titers in monkeys. Rhesus monkeys were immunized with a dual fusion virus vaccine formulated in HNS (alkaline buffer), AAHS or MAPA at 30 and 10 weeks at weeks 0 and 8. Serum samples were taken at week 12 and neutralized titers were assessed. Lines represent the geometric mean of the group.

圖11A-11B顯示在不同溫度下表現gH之CMV在漢克氏平衡鹽溶液(HBSS)中之穩定性。(A)在所示溫度下將存於 HBSS中之CMV試樣儲存4天,之後使用病毒進入分析量測CMV病毒穩定性。(B)使用病毒進入分析結果計算試樣之EC50值。指示因感染力完全損失而不能計算EC50。 Figures 11A-11B show the stability of CMV exhibiting gH at different temperatures in Hank's Balanced Salt Solution (HBSS). (A) CMV samples stored in HBSS were stored for 4 days at the indicated temperatures, after which CMV virus stability was measured using virus entry assay. (B) Calculate the EC50 value of the sample using the virus entry analysis results. * Indicates that EC50 cannot be calculated due to complete loss of infectivity.

圖12A-12B顯示在室溫下pH對表現gH之CMV之穩定性之效應。(A)在室溫下將存於具有不同pH之緩衝液中之CMV試樣儲存4天,之後使用病毒進入分析量測CMV病毒穩定性。(B)使用病毒進入分析結果計算試樣之EC50值。 Figures 12A-12B show the effect of pH on the stability of CMV exhibiting gH at room temperature. (A) CMV samples stored in buffers with different pH were stored for 4 days at room temperature, after which CMV virus stability was measured using virus entry assay. (B) Calculate the EC50 value of the sample using the virus entry analysis results.

圖13A-13B顯示尿素單獨或與氯化鈉組合對表現gH之CMV病毒穩定性之效應。(A)將2%尿素單獨或與150 mM NaCl組合添加至存於25 mM組胺酸緩衝液(pH 6)中之CMV中,且在室溫下保持4天,之後使用病毒進入分析量測CMV病毒穩定性。(B)使用病毒進入分析結果計算試樣之EC50值。 Figures 13A-13B show the effect of urea alone or in combination with sodium chloride on the stability of CMV virus expressing gH. (A) Add 2% urea alone or in combination with 150 mM NaCl to CMV in 25 mM histidine buffer (pH 6) and hold at room temperature for 4 days before using virus entry assay CMV virus stability. (B) Calculate the EC50 value of the sample using the virus entry analysis results.

圖14A-14B顯示離子強度對表現gH之CMV病毒穩定性之效應。(A)將增強濃度之NaCl(0 mM、75 mM、150 mM及320 mM NaCl)添加至存於25 mM組胺酸緩衝液(pH 6)中之CMV中,且在室溫下保持4天,之後使用病毒進入分析量測CMV病毒穩定性。(B)使用病毒進入分析結果計算試樣之EC50值。 Figures 14A-14B show the effect of ionic strength on the stability of CMV virus expressing gH. (A) Increasing concentrations of NaCl (0 mM, 75 mM, 150 mM, and 320 mM NaCl) were added to CMV in 25 mM histidine buffer (pH 6) and kept at room temperature for 4 days. Then, the virus was used to enter the assay to measure the stability of the CMV virus. (B) Calculate the EC50 value of the sample using the virus entry analysis results.

圖15顯示低溫保護劑對表現gH之CMV相對於冷凍-解凍循環之穩定性之效應。將所示低溫保護劑添加至存於25 mM組胺酸緩衝液(pH 6)中之CMV中且經受3次冷凍-解凍循環,之後使用病毒進入分析量測CMV病毒穩定性。使用病毒進入分析結果計算試樣之EC50值。 Figure 15 shows the effect of the cryoprotectant on the stability of the CMV exhibiting gH relative to the freeze-thaw cycle. The cryoprotectant shown was added to CMV in 25 mM histidine buffer (pH 6) and subjected to 3 freeze-thaw cycles, after which CMV virus stability was measured using virus entry assay. The EC50 value of the sample was calculated using the virus entry analysis results.

圖16A-16B顯示在小鼠免疫原性研究中儲存溫度對誘導CMV中和抗體之效應。在第0天對小鼠實施免疫且在第21天實施強化,之後在第28天抽血。使用ARPE-19細胞測試小鼠血清中針對表現gH之CMV之中和抗體。藉由非線性曲線擬合獲得NT50效價。(A)在免疫原性研究之前將CMV試樣在不同溫度下儲存3個月。(B)在解凍之後且在免疫原性研究之前將CMV試樣在不同溫度下儲存8小時。 Figures 16A-16B show the effect of storage temperature on induction of CMV neutralizing antibodies in a mouse immunogenicity study. Mice were immunized on day 0 and intensified on day 21, after which blood was drawn on day 28. CMV neutralizing antibodies against gH in mouse serum were tested using ARPE-19 cells. NT50 titers were obtained by nonlinear curve fitting. (A) CMV samples were stored at different temperatures for 3 months prior to immunogenicity studies. (B) CMV samples were stored at different temperatures for 8 hours after thawing and prior to immunogenicity studies.

<110> 美商默沙東公司 <110> American Merck Corporation

<120> 作為細胞巨大病毒疫苗之條件式複製CMV <120> Conditional replication CMV as a cellular giant virus vaccine

<130> 23109-US-PCT <130> 23109-US-PCT

<140> 101132064 <140> 101132064

<141> 2012-09-03 <141> 2012-09-03

<150> 61/532,667 <150> 61/532,667

<151> 2100-09-09 <151> 2100-09-09

<160> 14 <160> 14

<170> FastSEQ for Windows Version 4.0 <170> FastSEQ for Windows Version 4.0

<210> 1 <210> 1

<211> 598 <211> 598

<212> PRT <212> PRT

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 化學合成 <223> Chemical synthesis

<400> 1 <400> 1

<210> 2 <210> 2

<211> 2081 <211> 2081

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 化學合成 <223> Chemical synthesis

<400> 2 <400> 2

<210> 3 <210> 3

<211> 264 <211> 264

<212> PRT <212> PRT

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 化學合成 <223> Chemical synthesis

<400> 3 <400> 3

<210> 4 <210> 4

<211> 795 <211> 795

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 化學合成 <223> Chemical synthesis

<400> 4 <400> 4

<210> 5 <210> 5

<211> 775 <211> 775

<212> PRT <212> PRT

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 化學合成 <223> Chemical synthesis

<400> 5 <400> 5

<210> 6 <210> 6

<211> 2328 <211> 2328

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 化學合成 <223> Chemical synthesis

<400> 6 <400> 6

<210> 7 <210> 7

<211> 402 <211> 402

<212> PRT <212> PRT

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 化學合成 <223> Chemical synthesis

<400> 7 <400> 7

<210> 8 <210> 8

<211> 1209 <211> 1209

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 化學合成 <223> Chemical synthesis

<400> 8 <400> 8

<210> 9 <210> 9

<211> 693 <211> 693

<212> PRT <212> PRT

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 化學合成 <223> Chemical synthesis

<400> 9 <400> 9

<210> 10 <210> 10

<211> 107 <211> 107

<212> PRT <212> PRT

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 化學合成 <223> Chemical synthesis

<400> 10 <400> 10

<210> 11 <210> 11

<211> 2082 <211> 2082

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 化學合成 <223> Chemical synthesis

<400> 11 <400> 11

<210> 12 <210> 12

<211> 107 <211> 107

<212> PRT <212> PRT

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 化學合成 <223> Chemical synthesis

<400> 12 <400> 12

<210> 13 <210> 13

<211> 321 <211> 321

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 化學合成 <223> Chemical synthesis

<400> 13 <400> 13

<210> 14 <210> 14

<211> 230966 <211> 230966

<212> DNA <212> DNA

<213> 人工序列 <213> Artificial sequence

<220> <220>

<223> 化學合成 <223> Chemical synthesis

<400> 14 <400> 14

Claims (35)

一種條件式複製缺陷型細胞巨大病毒(CMV),其包含:(a)五聚gH複合物,其包含UL128、UL130、UL131、gH及gL;及(b)編碼必需蛋白IE1/2與去穩定蛋白間之第一融合蛋白及必需蛋白UL51與該去穩定蛋白間之第二融合蛋白之核酸,其中該去穩定蛋白為FK506-結合蛋白(FKBP)或FKBP衍生物,其包含胺基酸取代F36V及L106P,其中野生型IE1/2及UL51已不存在,且其中該CMV為減毒株,其因修復UL131基因中之突變而具有恢復之gH複合物表現。 A conditionally replicating defective cell giant virus (CMV) comprising: (a) a pentameric gH complex comprising UL128, UL130, UL131, gH and gL; and (b) encoding an essential protein IE1/2 and destabilizing a first fusion protein between proteins and a nucleic acid of a second fusion protein between the essential protein UL51 and the destabilizing protein, wherein the destabilizing protein is a FK506-binding protein (FKBP) or a FKBP derivative comprising an amino acid substituted F36V And L106P, wherein wild type IE1/2 and UL51 are no longer present, and wherein the CMV is an attenuated strain which has a restored gH complex expression due to repair of a mutation in the UL131 gene. 如請求項1之條件式複製缺陷型CMV,其中該FKBP衍生物係包含胺基酸取代F36V及L106P之FKBP。 The conditional replication defective CMV of claim 1, wherein the FKBP derivative comprises an amino acid substituted FKBP of F36V and L106P. 如請求項1或2之條件式複製缺陷型CMV,其中(a)該第一融合蛋白係SEQ ID NO:1;及(b)該第二融合蛋白係SEQ ID NO:3。 The conditional replication defective CMV of claim 1 or 2, wherein (a) the first fusion protein is SEQ ID NO: 1; and (b) the second fusion protein is SEQ ID NO: 3. 如請求項3之條件式複製缺陷型CMV,其中(a)該第一融合蛋白係由SEQ ID NO:2編碼;及(b)該第二融合蛋白係由SEQ ID NO:4編碼。 The conditional replication defective CMV of claim 3, wherein (a) the first fusion protein is encoded by SEQ ID NO: 2; and (b) the second fusion protein is encoded by SEQ ID NO: 4. 如請求項1之條件式複製缺陷型CMV,其中該CMV之基因組係SEQ ID NO:14。 The conditional replication defective CMV of claim 1 wherein the genome of the CMV is SEQ ID NO: 14. 如請求項1、2及5中任一項之條件式複製缺陷型CMV,其中該CMV為因修復UL131基因中之突變而具有恢復之gH複合物表現之AD 169。 The conditional replication-deficient CMV of any one of claims 1, 2, and 5, wherein the CMV is AD 169 having a restored gH complex manifested by repairing a mutation in the UL131 gene. 一種組合物,其包含如請求項1至6中任一項之條件式複製缺陷型CMV及醫藥上可接受之載劑。 A composition comprising the conditional replication-defective CMV of any one of claims 1 to 6 and a pharmaceutically acceptable carrier. 如請求項7之組合物,其進一步包含佐劑。 The composition of claim 7, which further comprises an adjuvant. 如請求項8之組合物,其中該佐劑係ISCOMATRIX®。 The composition of claim 8 wherein the adjuvant is ISCOMATRIX®. 一種如請求項7至9中任一項之組合物之用途,其用於製造用以在患者中誘導針對CMV之免疫反應之藥劑。 Use of a composition according to any one of claims 7 to 9 for the manufacture of a medicament for inducing an immune response against CMV in a patient. 如請求項10之用途,其中該免疫反應係患者中針對CMV感染之保護性免疫反應。 The use of claim 10, wherein the immune response is a protective immune response against CMV infection in a patient. 如請求項10或11之用途,其中該CMV感染係原發性感染、復發性感染或再感染(super-infection)。 The use of claim 10 or 11, wherein the CMV infection is a primary infection, a recurrent infection or a super-infection. 如請求項10或11之用途,其中該患者係人類。 The use of claim 10 or 11, wherein the patient is a human. 如請求項13之用途,其中該患者之免疫力減弱。 The use of claim 13 wherein the patient's immunity is reduced. 如請求項14之用途,其中該免疫力減弱之患者患有HIV或AIDS或係移植接受者。 The use of claim 14, wherein the immunocompromised patient has HIV or AIDS or a transplant recipient. 如請求項13之用途,其中該患者係育齡女性。 The use of claim 13, wherein the patient is a woman of childbearing age. 一種如請求項1至6中任一項之條件式複製缺陷型CMV之用途,其用於製造用以在患者中誘導針對CMV感染之保護性免疫反應之藥劑。 Use of a conditional replication-deficient CMV according to any one of claims 1 to 6 for the manufacture of a medicament for inducing a protective immune response against CMV infection in a patient. 如請求項1、2及5中任一項之條件式複製缺陷型CMV,其用於藥物中以治療患者之CMV感染。 The conditional replication-deficient CMV of any one of claims 1, 2, and 5 for use in a medicament for treating a CMV infection in a patient. 一種製備如請求項1至6中任一項之條件式複製缺陷型CMV之方法,其包含在Shield-1存在下在上皮細胞中繁殖重組CMV。 A method of producing a conditional replication-deficient CMV according to any one of claims 1 to 6, which comprises propagating recombinant CMV in epithelial cells in the presence of Shield-1. 如請求項19之方法,其中該等上皮細胞係人類色素視網 膜上皮細胞。 The human pigmentation network of the epithelial cell line Membrane epithelial cells. 如請求項20之方法,其中該等人類視網膜色素上皮細胞係以登錄號CRL-2302寄存於美國典型培養物保藏中心(American Type Culture Collection,ATCC)之ARPE-19細胞。 The method of claim 20, wherein the human retinal pigment epithelial cell line is deposited with ARPE-19 cells of the American Type Culture Collection (ATCC) under accession number CRL-2302. 如請求項19之方法,其中該Shield-1係以至少0.5μM之濃度存在。 The method of claim 19, wherein the Shield-1 is present at a concentration of at least 0.5 μM. 如請求項22之方法,其中該Shield-1係以至少2μM之濃度存在。 The method of claim 22, wherein the Shield-1 is present in a concentration of at least 2 μM. 一種製備如請求項7至9中任一項之組合物之方法,其包含:(a)在Shield-1添加之培養基中在上皮細胞中繁殖如請求項1至6中任一項之條件式複製缺陷型CMV;(b)自該培養基採集該條件式複製缺陷型CMV;(c)自該條件式複製缺陷型CMV實質上去除該Shield-1;及(d)將該醫藥上可接受之載劑添加至該條件式複製缺陷型CMV中。 A method of preparing a composition according to any one of claims 7 to 9 comprising: (a) breeding in an epithelial cell in a Shield-1 added medium, as in the conditional expression of any one of claims 1 to 6. Copying defective CMV; (b) collecting the conditional replication defective CMV from the culture medium; (c) substantially removing the Shield-1 from the conditional replication defective CMV; and (d) pharmaceutically acceptable A carrier is added to the conditional replication-defective CMV. 如請求項24之方法,其中該等上皮細胞係人類色素視網膜上皮細胞。 The method of claim 24, wherein the epithelial cells are human pigmentary retinal epithelial cells. 如請求項25之方法,其中該等人類視網膜色素上皮細胞係以登錄號CRL-2302寄存於美國典型培養物保藏中心(ATCC)之ARPE-19細胞。 The method of claim 25, wherein the human retinal pigment epithelial cell line is deposited with ARPE-19 cells of the American Type Culture Collection (ATCC) under accession number CRL-2302. 如請求項24之方法,其中在步驟(a)期間Shield-1係以至 少0.5μM之濃度存在於該培養基中。 The method of claim 24, wherein during the step (a), the Shield-1 is A concentration of 0.5 μM was present in the medium. 如請求項27之方法,其中在步驟(a)期間Shield-1係以至少2μM之濃度存在於該培養基中。 The method of claim 27, wherein the Shield-1 is present in the medium at a concentration of at least 2 μM during step (a). 如請求項24之方法,其中該去除係藉由超速離心或透析過濾。 The method of claim 24, wherein the removing is by ultracentrifugation or diafiltration. 如請求項24之方法,其中該去除使該Shield-1降至低於0.1μM。 The method of claim 24, wherein the removing reduces the Shield-1 to less than 0.1 μM. 一種組合物,其包含如請求項1至6中任一項之條件式複製缺陷型CMV於pH介於5至7之間之緩衝液中,該緩衝液包含:(a)介於15mM至35mM之間之組胺酸;及(b)介於100mM至200mM之間之NaCl。 A composition comprising the conditional replication-deficient CMV of any one of claims 1 to 6 in a buffer having a pH between 5 and 7, the buffer comprising: (a) between 15 mM and 35 mM Between the histidine; and (b) between 100 mM and 200 mM NaCl. 如請求項31之組合物,其進一步包含介於5%至15%之間之蔗糖。 The composition of claim 31, further comprising between 5% and 15% sucrose. 如請求項31至32中任一項之組合物,其中該緩衝液包含25mM組胺酸與150mM NaCl,pH為6。 The composition of any one of clauses 31 to 32, wherein the buffer comprises 25 mM histidine and 150 mM NaCl, pH 6. 如請求項33之組合物,其進一步包含9% w/v蔗糖。 The composition of claim 33, which further comprises 9% w/v sucrose. 一種SEQ ID NO:14之核酸。 A nucleic acid of SEQ ID NO: 14.
TW101132064A 2011-09-09 2012-09-03 A conditional replicating cytomegalovirus as a vaccine for cmv TWI570240B (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US201161532667P 2011-09-09 2011-09-09

Publications (2)

Publication Number Publication Date
TW201311898A TW201311898A (en) 2013-03-16
TWI570240B true TWI570240B (en) 2017-02-11

Family

ID=47832753

Family Applications (1)

Application Number Title Priority Date Filing Date
TW101132064A TWI570240B (en) 2011-09-09 2012-09-03 A conditional replicating cytomegalovirus as a vaccine for cmv

Country Status (33)

Country Link
US (1) US9546355B2 (en)
EP (3) EP3251700B1 (en)
JP (1) JP5926804B2 (en)
KR (1) KR101668163B1 (en)
CN (1) CN103889462B (en)
AR (1) AR087772A1 (en)
AU (1) AU2012304783B2 (en)
BR (1) BR112014005404A8 (en)
CA (1) CA2846870C (en)
CL (1) CL2014000541A1 (en)
CY (2) CY1119107T1 (en)
DK (2) DK2753364T3 (en)
ES (2) ES2633190T3 (en)
HR (2) HRP20171120T1 (en)
HU (2) HUE035322T2 (en)
IL (1) IL231272B (en)
IN (1) IN2014CN01809A (en)
LT (2) LT3251700T (en)
ME (2) ME03488B (en)
MX (1) MX353037B (en)
NZ (1) NZ622156A (en)
PE (1) PE20141525A1 (en)
PL (2) PL3251700T3 (en)
PT (2) PT3251700T (en)
RS (2) RS58891B1 (en)
RU (2) RU2670012C1 (en)
SG (1) SG11201400419YA (en)
SI (2) SI2753364T1 (en)
TR (1) TR201910644T4 (en)
TW (1) TWI570240B (en)
UA (1) UA114896C2 (en)
WO (1) WO2013036465A2 (en)
ZA (1) ZA201401708B (en)

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2016501023A (en) * 2012-12-04 2016-01-18 メルク・シャープ・アンド・ドーム・コーポレーションM Conditionally replicating viral vectors
MX2015016978A (en) * 2013-06-10 2016-04-25 Merck Sharp & Dohme Cmv neutralizing antigen binding proteins.
WO2016011293A1 (en) 2014-07-16 2016-01-21 Oregon Health & Science University Human cytomegalovirus comprising exogenous antigens
EP3048114A1 (en) * 2015-01-22 2016-07-27 Novartis AG Cytomegalovirus antigens and uses thereof
WO2017070613A1 (en) 2015-10-22 2017-04-27 Modernatx, Inc. Human cytomegalovirus vaccine
US10611800B2 (en) 2016-03-11 2020-04-07 Pfizer Inc. Human cytomegalovirus gB polypeptide
US11446398B2 (en) 2016-04-11 2022-09-20 Obsidian Therapeutics, Inc. Regulated biocircuit systems
WO2018075980A1 (en) 2016-10-21 2018-04-26 Modernatx, Inc. Human cytomegalovirus vaccine
BR112020008482A8 (en) * 2017-11-01 2023-01-31 Merck Sharp & Dohme LEUCU COMPOUND
US20210047626A1 (en) * 2018-04-24 2021-02-18 Merck Sharp & Dohme Corp. Scalable chromatography process for purification of human cytomegalovirus
US11629172B2 (en) 2018-12-21 2023-04-18 Pfizer Inc. Human cytomegalovirus gB polypeptide
US20220090005A1 (en) 2019-01-03 2022-03-24 Merck Sharp & Dohme Corp. Supplemented Serum-Containing Culture Medium for Enhanced Arpe-19 Growth and Human Cytomegalovirus Vaccine Production
CA3146798A1 (en) * 2019-07-12 2021-01-21 Kobenhavns Universitet Fusion toxin proteins for treatment of diseases related to cmv infections
US11857622B2 (en) 2020-06-21 2024-01-02 Pfizer Inc. Human cytomegalovirus GB polypeptide
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine
CN112481222B (en) * 2020-11-30 2024-01-02 中国医学科学院医学生物学研究所 Conditionally replicating recombinant herpes simplex virus, vaccine and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6251678B1 (en) * 1999-02-05 2001-06-26 Merck & Co., Inc. Human papillomavirus vaccine formulations
US20060110406A1 (en) * 2003-02-25 2006-05-25 Medimmune Vaccines, Inc. Refrigerator-temperature stable influenza vaccine compositions
US20090215169A1 (en) * 2007-02-09 2009-08-27 Wandless Thomas J Method for regulating protein function in cells using synthetic small molecules

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2429338T3 (en) 2002-12-23 2013-11-14 Vical Incorporated Codon-optimized polynucleotide-based vaccine against human cytomegalovirus infection
WO2004076645A2 (en) * 2003-02-27 2004-09-10 University Of Massachusetts Compositions and methods for cytomegalovirus treatment
NZ560929A (en) * 2005-02-18 2009-12-24 Novartis Vaccines & Diagnostic Immunogens from uropathogenic escherichia coli
EP2037959B1 (en) * 2006-06-07 2016-01-27 The Trustees Of Princeton University Cytomegalovirus surface protein complex for use in vaccines and as a drug target
US9439960B2 (en) * 2007-10-10 2016-09-13 The Trustees Of Princeton University Cytomegalovirus vaccines and methods of production
US8530636B2 (en) * 2008-05-07 2013-09-10 The Board Of Trustees Of The Leland Stanford Junior University Method for regulating protein function in cells in vivo using synthetic small molecules

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6251678B1 (en) * 1999-02-05 2001-06-26 Merck & Co., Inc. Human papillomavirus vaccine formulations
US20060110406A1 (en) * 2003-02-25 2006-05-25 Medimmune Vaccines, Inc. Refrigerator-temperature stable influenza vaccine compositions
US20090215169A1 (en) * 2007-02-09 2009-08-27 Wandless Thomas J Method for regulating protein function in cells using synthetic small molecules

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Glass M et al., "Conditional and reversible disruption of essential herpesvirus proteins", Nat Methods. 2009 Aug 6(8):577-9. Epub 2009 Jul 5. *
Mark A. Jacobsona et al., "Safety and immunogenicity of Towne cytomegalovirus vaccine with or without adjuvant recombinant interleukin-12", Vaccine. Volume 24, Issue 25, 19 June 2006, Pages 5311–5319. *
Sarah Straschewski et al., "Protein pUL128 of Human Cytomegalovirus Is Necessary for Monocyte Infection and Blocking of Migration", JOURNAL OF VIROLOGY, May 2011, p. 5150–5158. *

Also Published As

Publication number Publication date
CL2014000541A1 (en) 2014-08-22
IL231272A0 (en) 2014-04-30
SG11201400419YA (en) 2014-04-28
ME03488B (en) 2020-01-20
UA114896C2 (en) 2017-08-28
WO2013036465A3 (en) 2013-05-10
CN103889462A (en) 2014-06-25
PE20141525A1 (en) 2014-11-12
EP3251700A1 (en) 2017-12-06
KR20140057654A (en) 2014-05-13
SI3251700T1 (en) 2019-08-30
HUE035322T2 (en) 2018-05-02
HRP20191111T1 (en) 2019-09-20
MX2014002815A (en) 2014-04-10
RU2670012C1 (en) 2018-10-17
US20140220062A1 (en) 2014-08-07
WO2013036465A2 (en) 2013-03-14
TR201910644T4 (en) 2019-08-21
BR112014005404A2 (en) 2017-03-28
EP3572096A1 (en) 2019-11-27
LT2753364T (en) 2017-07-25
CY1121863T1 (en) 2020-07-31
JP2014527809A (en) 2014-10-23
RU2611198C2 (en) 2017-02-21
BR112014005404A8 (en) 2023-04-11
AU2012304783A1 (en) 2014-03-13
ES2737852T3 (en) 2020-01-16
CY1119107T1 (en) 2018-02-14
LT3251700T (en) 2019-08-12
ME02831B (en) 2018-01-20
HRP20171120T1 (en) 2017-10-06
PT3251700T (en) 2019-07-25
RS58891B1 (en) 2019-08-30
CA2846870A1 (en) 2013-03-14
NZ622156A (en) 2015-12-24
HUE045115T2 (en) 2019-12-30
PT2753364T (en) 2017-07-18
KR101668163B1 (en) 2016-10-20
EP3251700B1 (en) 2019-05-22
PL2753364T3 (en) 2017-11-30
EP2753364A2 (en) 2014-07-16
MX353037B (en) 2017-12-18
RS56261B1 (en) 2017-11-30
AR087772A1 (en) 2014-04-16
ES2633190T3 (en) 2017-09-19
JP5926804B2 (en) 2016-05-25
AU2012304783B2 (en) 2016-03-10
CN103889462B (en) 2016-12-28
DK3251700T3 (en) 2019-08-26
IN2014CN01809A (en) 2015-05-29
ZA201401708B (en) 2014-11-26
EP2753364B1 (en) 2017-05-24
RU2014113921A (en) 2015-10-20
IL231272B (en) 2019-01-31
PL3251700T3 (en) 2019-09-30
US9546355B2 (en) 2017-01-17
CA2846870C (en) 2017-10-03
DK2753364T3 (en) 2017-08-28
TW201311898A (en) 2013-03-16
SI2753364T1 (en) 2017-08-31
EP2753364A4 (en) 2015-04-08

Similar Documents

Publication Publication Date Title
TWI570240B (en) A conditional replicating cytomegalovirus as a vaccine for cmv
KR100372934B1 (en) Virus Defect Vaccines Generated by Transgenic Complementary Cell Lines
US20080044384A1 (en) Recombinant Human Cytomegalovirus And Vaccines Comprising Heterologous Antigens
RU2491340C2 (en) Cytomegalovirus vaccine and method for preparing it
US20100272752A1 (en) Vaccine for viruses that cause persistent or latent infections
US20150307850A1 (en) Conditional replicating viral vectors
US20230277654A1 (en) Stable formulations of cytomegalovirus
Tai et al. In vitro characterization of felid herpesvirus 1 (FHV-1) mutants generated by recombineering in a recombinant BAC vector
Gray Recombinant varicella-zoster virus vaccines as platforms for expression of foreign antigens
US20230372473A1 (en) Vaccine compositions
Stanfield et al. Rational Design of Live-Attenuated Vaccines against Herpes Simplex Viruses. Viruses 2021, 13, 1637
WO2023064612A2 (en) Pharmaceutical compositions for delivery of viral antigens and related methods